Compounds and compositions and uses thereof

Compounds of formula I: are disclosed, as are pharmaceutical compositions containing such compounds. Methods of treating neurological or psychiatric diseases and disorders in a subject in need thereof are also disclosed.

Skip to: Description  ·  Claims  ·  References Cited  · Patent History  ·  Patent History
Description
FIELD OF THE INVENTION

The invention relates to isochroman-4-amines, benzooxepinamines and benzooxacinamines as well as pharmaceutical compositions containing such compounds. The compounds and pharmaceutical compositions are useful for treating neurological or psychiatric diseases and disorders in a subject in need thereof. Such diseases and disorders include, for example, depression, bipolar disorder, pain, schizophrenia, obsessive compulsive disorder, addiction, social disorder, attention deficit hyperactivity disorder, an anxiety disorder, autism, a cognitive impairment, or a neuropsychiatric symptom such as apathy, depression, anxiety, psychosis, aggression, agitation, impulse control disorders, and sleep disorders in neurological disorders such as Alzheimer's and Parkinson's diseases.

All publications, patents, patent applications, and other references cited in this application are incorporated herein by reference in their entirety for all purposes and to the same extent as if each individual publication, patent, patent application or other reference was specifically and individually indicated to be incorporated by reference in its entirety for all purposes. Citation of a reference herein shall not be construed as an admission that such is prior art to the present invention.

BACKGROUND OF THE INVENTION

Central nervous system diseases and disorders affect a wide range of the population with differing severity. Neurological and psychiatric diseases and disorders include major depression, schizophrenia, bipolar disorder, obsessive compulsive disorder (OCD), panic disorder, and posttraumatic stress disorder (PTSD), among others. These diseases and disorders affect a person's thoughts, mood, behavior and social interactions and can significantly impair daily functioning. See, e.g., Diagnostic and Statistical Manual of Mental Disorders, 4th Ed., American Psychiatric Association (2000) (“DSM-IV-TR”); Diagnostic and Statistical Manual of Mental Disorders, 5th Ed., American Psychiatric Association (2013) (“DSM-5”).

While medications exist for some aspects of these diseases and disorders, there remains a need for effective treatments for various neurological and psychiatric diseases and disorders, including mood disorders such as bipolar and related disorders, psychosis and schizophrenia. For example, while mood stabilizers such as lithium and valproate, antidepressants and antipsychotic drugs are used to treat mood disorders, more effective medications are necessary. And current antipsychotics may be successful in treating the positive symptoms of schizophrenia but fare less well for the negative and cognitive symptoms. Additionally, current antidepressants are typically effective only for a proportion of subjects suffering from depression. Furthermore, despite the fact that the behavioral and psychiatric symptoms of neurological disease such as Parkinson's disease and Alzheimer's disease are major reasons for the institutionalization of subjects, few drugs exist to treat them.

SUMMARY OF THE INVENTION

In one aspect, the invention relates to method for treating a neurological or psychiatric disease or disorder comprising administering to a subject in need thereof an effective amount of a compound of formula I:

or a pharmaceutically acceptable salt thereof,

wherein:

q1 is 0 or 1,

q2 is 0 or 1, and

the sum of q1 plus q2 is 0 or 1;

R1, R2 and R3 are chosen independently from H and an aliphatic (C1-C8)hydrocarbyl optionally substituted with one or more groups independently selected from halogen, —C≡C—, hydroxyl, (C1-C6)alkoxy, amino, (C1-C6)alkylamino and di(C1-C6)alkylamino;

R4a, R4b, R7a, and R7b are chosen independently from H, halogen, (C1-C6)alkyl, (C1-C6)haloalkyl, and (C1-C6)alkoxy;

R5a, R5b, R6a and R6b are chosen independently from H, halogen, (C1-C6)alkyl and (C1-C6)haloalkyl;

R8, R9, R10 and R11 are chosen independently from H, halogen, (C1-C8)hydrocarbyl, cyano, —CF3, (C1-C6)haloalkyl, hydroxyl, (C1-C6)alkoxy, aminocarbonyl, (C1-C6)alkylaminocarbonyl, di(C1-C6)alkylaminocarbonyl, (C1-C6)acyl, (C1-C6)haloalkoxy, hydroxy(C1-C6)alkyl, carboxy, (C1-C6)alkoxycarbonyl, acetoxy, nitro, amino, (C1-C6)alkylamino, di(C1-C6)alkylamino, aminosulfonyl, benzyl, aryl, heteroaryl, phenoxy, —OC(O)N(alkyl)2 or benzyloxy;

wherein said benzyl, aryl, heteroaryl, phenoxy or benzyloxy are optionally substituted with one or more substituents independently selected from halogen, (C1-C6)alkyl, cyano, (C1-C6)haloalkyl, hydroxyl, (C1-C6)alkoxy, aminocarbonyl, (C1-C6)alkylaminocarbonyl, di(C1-C6)alkylaminocarbonyl, (C1-C6)acyl, (C1-C6)haloalkoxy, hydroxy(C1-C6)alkyl, carboxy, (C1-C6)alkoxycarbonyl, acetoxy, nitro, amino, (C1-C6)alkylamino, and di(C1-C6)alkylamino;

and further wherein:

any two of R8, R9, R10 and R11 attached to adjacent carbon atoms, together with the carbon atoms to which they are attached, may form a 5 to 8-membered carbocycle or heterocycle which is optionally substituted with one or more substituents independently selected from halogen, hydroxyl, amino, (C1-C6)alkylamino, di(C1-C6)alkylamino, (C1-C6)alkyl, (C1-C6)haloalkyl, (C1-C6)alkoxy, and (C1-C6)haloalkoxy.

In another aspect the invention relates to pharmaceutical compositions comprising compounds of formula I above, wherein at least one of R1, R2, R3, R4a, R4b, R5a, R5b, R6a, R6b, R7a, R7b, R8, R9, R10 and R11 is other than hydrogen.

In another aspect, the invention relates to chemical compounds of formula I, with the provisos that:

when q1 and q2 are both zero and NR2R3 is NHCH3 or N(CH3)2, then at least one of R1, R4a, R4b, R8, R9, R10 and R11 must be other than hydrogen or methoxy, and

when q1 and q2 are both zero and NR2R3 is NH2, then one of the following four conditions must be met:

at least two of R8, R9, R10 and R11 must be other than hydrogen, or

at least one of R8, R9, R10 and R11 must be phenyl, or

at least one of R4a and R4b, must be other than hydrogen, or

at least one of R7a and R7b must be methyl and the other of R7a and R7b must be other than methyl, and

when q1 or q2 is one and NR2R3 is NH2, then the following species are excluded:

BRIEF DESCRIPTION OF THE DRAWINGS

FIG. 1 shows the effect of representative compounds of the invention on treatment on DA release in the PFC.

FIG. 2 shows the effect the effect of representative compounds of the invention on treatment on 5-HT release in the PFC.

FIG. 3 shows the effect of representative compounds of the invention on treatment on NE release in the PFC.

FIG. 4 shows the effect of representative compounds of the invention on treatment on ACh release in the PFC

FIG. 5 shows the effect of representative compounds of the invention on treatment on DA release in the NAcc.

FIG. 6 shows the effect of representative compounds of the invention on treatment on 5-HT release in the NAcc.

FIG. 7 shows the effect of representative compounds of the invention on treatment on NE release in the NAcc.

FIG. 8 shows the effect of representative compounds of the invention on treatment on ACh release in the NAcc.

DETAILED DESCRIPTION OF THE INVENTION

The methods of the invention relate to the use of compounds and compositions of Formula I above to treat neurological or psychiatric diseases and disorders or impairments. In some embodiments, the neurological or psychiatric disease or disorder is depression, bipolar disorder, pain, schizophrenia, obsessive compulsive disorder, addiction, social disorder, attention deficit hyperactivity disorder, an anxiety disorder, autism or cognitive impairments. In one embodiment, the disorder is depression, particularly treatment-resistant depression (TRD), major depressive disorder (MDD), unipolar depression, bipolar depression or depression associated with another disease or disorder.

In one embodiment relating to the use of compounds of formula I in these methods, any two of R8, R9, R10 and R11 attached to adjacent carbon atoms, together with the carbon atoms to which they are attached, may form a 5 to 8-membered carbocycle or heterocycle which is optionally substituted with one or more substituents independently selected from halogen, hydroxyl, amino, (C1-C6)alkylamino, di(C1-C6)alkylamino, (C1-C6)alkyl, (C1-C6)haloalkyl, (C1-C6)alkoxy, and (C1-C6)haloalkoxy. In one such embodiment, any two of R8, R9, R10 and R11 form a phenyl ring, which may be substituted as described.

In another embodiment relating to the use of compounds of formula I in these methods, there are four possibilities: (a) at least one of R8, R9, and R11 is chosen from halogen, methyl and ethyl; or (b) R10 is methoxy; or (c) R8, R9, R10, R11, R7a, and R7b are hydrogen and at least one of R4a and R4b is methyl, or (d) R8 and R9, together with the carbon atoms to which they are attached, form a 6-membered carbocycle or 7-membered bridged carbocycle. In any of these four possibilities, when q1 and q2 are both zero, R4a may be hydrogen and R4b may be hydrogen or methyl, and R2 and R3 may be hydrogen. In any of these four possibilities, when q1 is one, R2 and R3 may be hydrogen or methyl.

In another embodiment relating to the use of compounds of formula I in these methods, exemplary compounds are those in which R2 and R3 are hydrogen, q1 and q2 are both zero, and (a) both of R7a and R7b are methyl, or (b) both of R8 and R9 are methyl, or (c) R9 is methoxy and at least one of R4a and R4b is methyl, or (d) R11 is propyl.

In another embodiment relating to the use of compounds of formula I in these methods, exemplary compounds are those in which R4a, R4b, R5a, R5b, R6a, R6b, R7a, and R7b are all hydrogen, and (a) q1 is one and R1, R2, R3, R8, R10, and R11 are hydrogen, or (b) q1 and q2 are both zero, R10 and R11 are hydrogen, or (c) q1 and q2 are both zero, R8, R9, and R10 are hydrogen and R11 is —(C1-C3)alkyl.

In another embodiment relating to the use of compounds of formula I in these methods, exemplary compounds are those in which R1, R4a, R4b, R5a, R5b, R6a, R6b, R7a, and R7b are all hydrogen, and (a) q1 is one, R2 and R3 are hydrogen or methyl, and R10 and R11 are hydrogen or methyl, or (b) q1 and q2 are both zero and R8, R9, R10 and R11 are chosen from hydrogen, methyl, methoxy and chloro.

In a pharmaceutical composition aspect, the invention relates to compositions comprising a pharmaceutically acceptable carrier and a compound of formula I above, wherein at least one of R1, R2, R3 R4a, R4b, R5a, R5b, R6a, R6b, R7a, R7b, R8, R9, R10 and R11 is other than hydrogen.

In a chemical compound aspect, the invention relates to compounds of formula I:

or a pharmaceutically acceptable salt thereof,

wherein:

q1 is 0 or 1,

q2 is 0 or 1, and

the sum of q1 plus q2 is 0 or 1;

R1, R2 and R3 are chosen independently from H and an aliphatic (C1-C8)hydrocarbyl optionally substituted with one or more groups independently selected from halogen, —C≡C—, hydroxyl, (C1-C6)alkoxy, amino, (C1-C6)alkylamino and di(C1-C6)alkylamino;

R4a, R4b, R7a, and R7b are chosen independently from H, halogen, (C1-C6)alkyl, (C1-C6)haloalkyl, and (C1-C6)alkoxy;

R5a, R5b, R6a and R6b are chosen independently from H, halogen, (C1-C6)alkyl and (C1-C6)haloalkyl;

R8, R9, R10 and R11 are chosen independently from H, halogen, (C1-C8)hydrocarbyl, cyano, —CF3, (C1-C6)haloalkyl, hydroxyl, (C1-C6)alkoxy, aminocarbonyl, (C1-C6)alkylaminocarbonyl, di(C1-C6)alkylaminocarbonyl, (C1-C6)acyl, (C1-C6)haloalkoxy, hydroxy(C1-C6)alkyl, carboxy, (C1-C6)alkoxycarbonyl, acetoxy, nitro, amino, (C1-C6)alkylamino, di(C1-C6)alkylamino, aminosulfonyl, benzyl, aryl, heteroaryl, phenoxy, —OC(O)N(alkyl)2 or benzyloxy;

wherein said benzyl, aryl, heteroaryl, phenoxy or benzyloxy are optionally substituted with one or more substituents independently selected from halogen, (C1-C6)alkyl, cyano, (C1-C6)haloalkyl, hydroxyl, (C1-C6)alkoxy, aminocarbonyl, (C1-C6)alkylaminocarbonyl, di(C1-C6)alkylaminocarbonyl, (C1-C6)acyl, (C1-C6)haloalkoxy, hydroxy(C1-C6)alkyl, carboxy, (C1-C6)alkoxycarbonyl, acetoxy, nitro, amino, (C1-C6)alkylamino, and di(C1-C6)alkylamino;

and further wherein:

any two of R8, R9, R10 and R11 attached to adjacent carbon atoms, together with the carbon atoms to which they are attached, may form a 5 to 8-membered carbocycle or heterocycle which is optionally substituted with one or more substituents independently selected from halogen, hydroxyl, amino, (C1-C6)alkylamino, di(C1-C6)alkylamino, (C1-C6)alkyl, (C1-C6)haloalkyl, (C1-C6)alkoxy, and (C1-C6)haloalkoxy;

with the provisos that,

when q1 and q2 are both zero and NR2R3 is NHCH3 or N(CH3)2, then at least one of R1, R4a, R4b, R8, R9, R10 and R11 must be other than hydrogen or methoxy; or

when q1 and q2 are both zero and NR2R3 is NH2, then one of the following four conditions must be met:

    • at least two of R8, R9, R10 and R11 must be other than hydrogen, or
    • at least one of R8, R9, R10 and R11 must be phenyl, or
    • at least one of R8, R9, R10 and R11 must be phenyl, or
    • at least one of R4a and R4b must be other than hydrogen, or
    • at least one of R7a and R7b must be methyl and the other of R7a and R7b must be other than methyl, and
    • when q1 or q2 is one, then the following species are excluded:

In one embodiment relating to the compounds, q1 and q2 are both zero, and the compounds are chroman-1-amines of formula II:

In another embodiment relating to the compounds, q1 is one, and the compounds are 1,3,4,5-tetrahydrobenzo[c]oxepin-5-amines of formula III:

In another embodiment relating to the compounds, q2 is one, and the compounds are 1,2,4,5-tetrahydrobenzo[d]oxepin-1-amines of formula IV:

In some embodiments, R4a, R4b, R7a, and R7b are hydrogen.

In some embodiments, R7 and R8 form a 5 to 8-membered carbocycle or heterocycle which is optionally substituted with one or more substituents independently selected from halogen, hydroxyl, amino, (C1-C6)alkylamino, di(C1-C6)alkylamino, (C1-C6)alkyl, (C1-C6)haloalkyl, (C1-C6)alkoxy, and (C1-C6)haloalkoxy. In some embodiments, the carbocycle or heterocycle may be chosen from phenyl, pyridine, cyclohexene, thiazole, bicycloheptene, and dihydropyran.

In some embodiments, R9 and R10 form a 5 to 8-membered carbocycle or heterocycle which is optionally substituted with one or more substituents independently selected from halogen, hydroxyl, amino, (C1-C6)alkylamino, di(C1-C6)alkylamino, (C1-C6)alkyl, (C1-C6)haloalkyl, (C1-C6)alkoxy, and (C1-C6)haloalkoxy. In some embodiments, the carbocycle or heterocycle may be chosen from phenyl, pyridine, cyclohexene, thiazole, bicycloheptene, and dihydropyran.

In some embodiments, R9 is a phenyl which is unsubstituted or substituted with 1 to 4 substituents independently selected from halo, —OH, —NH2, methyl, ethyl, methoxy, trifluoromethyl and trifluoromethoxy; in others, R9 is a 5- or 6-membered heteroaryl which is unsubstituted or substituted with 1 to 4 substituents independently selected from halo, —OH, —NH2, methyl, ethyl, methoxy, trifluoromethyl and trifluoromethoxy.

In some embodiments, two of R8, R9, R10 and R11 are chosen from methyl and chloro and the other two are hydrogen.

In some embodiments, R2 is hydrogen and R3 is chosen from methyl, ethyl, propyl, isopropyl, and hydroxyethyl.

In some embodiments, R1 is hydrogen or methyl; in some embodiments R1 is hydrogen.

In some embodiments, R4a and R4b are chosen independently from hydrogen and (C1-C6)alkyl.

In some embodiments of tetrahydrobenzo[c]oxepin-5-amines of formula III or 1,2,4,5-tetrahydrobenzo[d]oxepin-1-amines of formula IV, R8 and R9 form a 5 to 8-membered carbocycle or heterocycle which is optionally substituted with one or more substituents independently selected from halogen, hydroxyl, amino, (C1-C6)alkylamino, di(C1-C6)alkylamino, (C1-C6)alkyl, (C1-C6)haloalkyl, (C1-C6)alkoxy, and (C1-C6)haloalkoxy. In particular embodiments, the carbocycle is phenyl, which may be substituted as described.

In some embodiments of tetrahydrobenzo[c]oxepin-5-amines of formula III or 1,2,4,5-tetrahydrobenzo[d]oxepin-1-amines of formula IV, R4a and R4b are chosen from hydrogen and methyl.

In some embodiments of tetrahydrobenzo[c]oxepin-5-amines of formula III or 1,2,4,5-tetrahydrobenzo[d]oxepin-1-amines of formula IV, R2 is hydrogen and R3 is hydrogen or methyl.

In some embodiments of chroman-1-amines of formula II, tetrahydrobenzo[c]oxepin-5-amines of formula III or 1,2,4,5-tetrahydrobenzo[d]oxepin-1-amines of formula IV, one or two of R8, R9, R10 and R11 are chosen from methyl and chloro and the remaining two or three of R8, R9, R10 and R11 are hydrogen.

In one embodiment, provided are compounds of formula (I) which are greater than 90% enantiomerically pure. In another embodiment, provided are compounds of formula I which are greater than 95% enantiomerically pure.

In one embodiment, provided is a compound according to formula I, wherein said compound is:


or a pharmaceutically acceptable salt thereof.

In one embodiment, provided are compounds according to formula I wherein said compound is:


or a pharmaceutically acceptable salt thereof.

In another embodiment, provided are compounds according to formula I wherein said compound is:


or a pharmaceutically acceptable salt thereof.

In another embodiment, provided are compounds according to formula I wherein said compound is:


and has greater than 90% enantiomeric purity. In another embodiment, said compound has greater than 95% enantiomeric purity.

Compounds and Compositions and Definitions

Compounds and compositions of this invention include those described generally above, and are further illustrated by the classes, subclasses, and species disclosed herein. As used herein, the following definitions shall apply unless otherwise indicated. A comprehensive list of abbreviations utilized by organic chemists (i.e. persons of ordinary skill in the art) appears in the first issue of each volume of the Journal of Organic Chemistry. The definitions therein, which are typically presented in a table entitled “Standard List of Abbreviations” are the definitions used herein.

The graphic representations of racemic, ambiscalemic and scalemic or enantiomerically pure compounds used herein are a modified version of the denotations taken from Maehr J. Chem. Ed. 62, 114-120 (1985): simple lines provide no information about stereochemistry and convey only connectivity; solid and broken wedges are used to denote the absolute configuration of a chiral element; solid and broken bold lines are geometric descriptors indicating the relative configuration shown but not necessarily denoting racemic character; and wedge outlines and dotted or broken lines denote enantiomerically pure compounds of the indicated relative stereochemistry of indeterminate absolute configuration. For example, the graphic representation


indicates a trans relationship between the two chiral centers, that is, either or both of the two representations below:


in any ratio, from pure enantiomers to racemates, while the representation:


indicates a single enantiomer with the absolute configuration depicted, e.g., ((R)-1-((R)-5-fluoroisochroman-1-yl)ethan-1-amine in the illustration above. Further, the open wedge/broken line depiction as shown in the example below:


represents a single enantiomer of the indicated relative stereochemistry of an undetermined absolute configuration. In the text describing the stereochemistry of the examples, the convention of Chemical Abstracts is used. Thus “(R)-1-((R)-5-rel- . . . ” indicates that the two chiral centers are in that relative relationship, which would be depicted in a structural diagram by solid bold and dashed lines, whereas “(R)-1-((R)-5- . . . ” without the “rel” indicates a single enantiomer of that absolute configuration, which would be depicted in a structural diagram by solid and broken wedges.

The “enantiomeric excess” or “% enantiomeric excess” of a composition can be calculated using the equation shown below. In the example shown below, a composition contains 90% of one enantiomer, e.g., the S enantiomer, and 10% of the other enantiomer, e.g., the R enantiomer. ee=(90-10)/100=80%.

Thus, a composition containing 90% of one enantiomer and 10% of the other enantiomer is said to have an enantiomeric excess of 80%. Some compositions described herein contain an enantiomeric excess of at least about 50%, 75%, 90%, 95%, or 99% of the S enantiomer. In other words, the compositions contain an enantiomeric excess of the S enantiomer over the R enantiomer. In other embodiments, some compositions described herein contain an enantiomeric excess of at least about 50%, 75%, 90%, 95%, or 99% of the R enantiomer. In other words, the compositions contain an enantiomeric excess of the R enantiomer over the S enantiomer.

For instance, an isomer/enantiomer can, in some embodiments, be provided substantially free of the corresponding enantiomer, and can also be referred to as “optically enriched,” “enantiomerically enriched,” “enantiomerically pure” and “non-racemic,” as used interchangeably herein. These terms refer to compositions in which the percent by weight of one enantiomer is greater than the amount of that one enantiomer in a control mixture of the racemic composition (e.g., greater than 1:1 by weight). For example, an enantiomerically enriched preparation of the S enantiomer, means a preparation of the compound having greater than about 50% by weight of the S enantiomer relative to the R enantiomer, such as at least about 75% by weight, further such as at least about 80% by weight. In some embodiments, the enrichment can be much greater than about 80% by weight, providing a “substantially enantiomerically enriched,” “substantially enantiomerically pure” or a “substantially non-racemic” preparation, which refers to preparations of compositions which have at least about 85% by weight of one enantiomer relative to other enantiomer, such as at least about 90% by weight, and further such as at least 95% by weight. In certain embodiments, the compound provided herein is made up of at least about 90% by weight of one enantiomer. In other embodiments, the compound is made up of at least about 95%, 98%, or 99% by weight of one enantiomer.

In some embodiments, the compound is a racemic mixture of (S)- and (R)-isomers. In other embodiments, provided herein is a mixture of compounds wherein individual compounds of the mixture exist predominately in an (S)- or (R)-isomeric configuration. For example, the compound mixture has an (S)-enantiomeric excess of greater than about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, about 96%, about 97%, about 98%, about 99%, about 99.5%, or more. In other embodiments, the compound mixture has an (S)-enantiomeric excess of greater than about 55% to about 99.5%, greater than about 60% to about 99.5%, greater than about 65% to about 99.5%, greater than about 70% to about 99.5%, greater than about 75% to about 99.5%, greater than about 80% to about 99.5%, greater than about 85% to about 99.5%, greater than about 90% to about 99.5%, greater than about 95% to about 99.5%, greater than about 96% to about 99.5%, greater than about 97% to about 99.5%, greater than about 98% to greater than about 99.5%, greater than about 99% to about 99.5%, or more.

In other embodiments, the compound mixture has an (R)-enantiomeric purity of greater than about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, about 96%, about 97%, about 98%, about 99%, about 99.5% or more. In some other embodiments, the compound mixture has an (R)-enantiomeric excess of greater than about 55% to about 99.5%, greater than about 60% to about 99.5%, greater than about 65% to about 99.5%, greater than about 70% to about 99.5%, greater than about 75% to about 99.5%, greater than about 80% to about 99.5%, greater than about 85% to about 99.5%, greater than about 90% to about 99.5%, greater than about 95% to about 99.5%, greater than about 96% to about 99.5%, greater than about 97% to about 99.5%, greater than about 98% to greater than about 99.5%, greater than about 99% to about 99.5% or more.

In other embodiments, the compound mixture contains identical chemical entities except for their stereochemical orientations, namely (S)- or (R)-isomers. For example, if a compound disclosed herein has —CH(R)— unit, and R is not hydrogen, then the —CH(R)— is in an (S)- or (R)-stereochemical orientation for each of the identical chemical entities. In some embodiments, the mixture of identical chemical entities is a racemic mixture of (S)- and (R)-isomers. In another embodiment, the mixture of the identical chemical entities (except for their stereochemical orientations), contain predominately (S)-isomers or predominately (R)-isomers. For example, the (S)-isomers in the mixture of identical chemical entities are present at about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, about 96%, about 97%, about 98%, about 99%, about 99.5%, or more, relative to the (R)-isomers. In some embodiments, the (S)-isomers in the mixture of identical chemical entities are present at an (S)-enantiomeric excess of greater than about 55% to about 99.5%, greater than about 60% to about 99.5%, greater than about 65% to about 99.5%, greater than about 70% to about 99.5%, greater than about 75% to about 99.5%, greater than about 80% to about 99.5%, greater than about 85% to about 99.5%, greater than about 90% to about 99.5%, greater than about 95% to about 99.5%, greater than about 96% to about 99.5%, greater than about 97% to about 99.5%, greater than about 98% to greater than about 99.5%, greater than about 99% to about 99.5% or more.

In another embodiment, the (R)-isomers in the mixture of identical chemical entities (except for their stereochemical orientations), are present at about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, about 96%, about 97%, about 98%, about 99%, about 99.5%, or more, relative to the (S)-isomers. In some embodiments, the (R)-isomers in the mixture of identical chemical entities (except for their stereochemical orientations), are present at a (R)-enantiomeric excess greater than about 55% to about 99.5%, greater than about 60% to about 99.5%, greater than about 65% to about 99.5%, greater than about 70% to about 99.5%, greater than about 75% to about 99.5%, greater than about 80% to about 99.5%, greater than about 85% to about 99.5%, greater than about 90% to about 99.5%, greater than about 95% to about 99.5%, greater than about 96% to about 99.5%, greater than about 97% to about 99.5%, greater than about 98% to greater than about 99.5%, greater than about 99% to about 99.5%, or more.

Hydrocarbyl refers to any substituent comprised of hydrogen and carbon as the only elemental constituents. C1 to C20 hydrocarbon includes, for example, alkyl, cycloalkyl, polycycloalkyl, alkenyl, alkynyl, aryl and combinations thereof. Examples include benzyl, phenethyl, cyclohexylmethyl, adamantyl, camphoryl and naphthylethyl. Aromatic hydrocarbons include benzene (phenyl), naphthalene (naphthyl), anthracene, etc. Aliphatic hydrocarbons are hydrocarbons that are not aromatic; they may be saturated or unsaturated, cyclic, linear or branched, or combinations thereof. Aliphatic hydrocarbons include, for example, alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, cycloalkynyl, and combinations thereof. Non-limiting examples of aliphatic hydrocarbons include isopropyl, 2-butenyl, 2-butynyl, cyclopentyl, cyclopropylmethyl, norbornyl, and the like.

Unless otherwise specified, alkyl (or alkylene) is intended to include linear or branched saturated hydrocarbon structures and combinations thereof. Alkyl refers to alkyl groups from 1 to 20 carbon atoms, preferably 1 to 10 carbon atoms, more preferably 1 to 6 carbon atoms. Examples of alkyl groups include methyl, ethyl, propyl, isopropyl, n-butyl, s-butyl, t-butyl and the like.

Cycloalkyl is a subset of hydrocarbon and includes cyclic hydrocarbon groups of from 3 to 8 carbon atoms. Examples of cycloalkyl groups include cyclopropyl, cyclobutyl, cyclopentyl, norbornyl and the like.

Unless otherwise specified, the term “carbocycle” is intended to include ring systems in which the ring atoms are all carbon but of any oxidation state. Thus (C3-C10) carbocycle refers to both non-aromatic and aromatic systems, including such systems as cyclopropane, benzene (phenyl) and cyclohexene; (C8-C12) carbopolycycle refers to such systems as norbornane, decalin, indane and naphthalene. Carbocycle, if not otherwise limited, refers to monocycles, bicycles and polycycles, including bridged structures.

Heterocycle means an aliphatic or aromatic carbocycle residue in which from one to four carbons is replaced by a heteroatom selected from the group consisting of N, O, and S. The nitrogen and sulfur heteroatoms may optionally be oxidized, and the nitrogen heteroatom may optionally be quaternized. Unless otherwise specified, a heterocycle may be non-aromatic (heteroaliphatic) or aromatic (heteroaryl). Heterocycle, if not otherwise limited, refers to monocycles, bicycles and polycycles, including bridged structures. Examples of heterocycles include, but are not limited to, pyrrolidine, pyrazole, pyrrole, indole, quinoline, isoquinoline, tetrahydroisoquinoline, benzofuran, benzodioxan, benzodioxole (commonly referred to as methylenedioxyphenyl, when occurring as a substituent), tetrazole, morpholine, thiazole, pyridine, pyridazine, pyrimidine, thiophene, furan, oxazole, oxazoline, isoxazole, atrophine, dioxane, tetrahydrofuran and the like. Examples of heterocyclyl residues include piperazinyl, piperidinyl, pyrazolidinyl, imidazolyl, imidazolinyl, imidazolidinyl, pyrazinyl, oxazolidinyl, isoxazolidinyl, thiazolidinyl, isothiazolyl, quinuclidinyl, isothiazolidinyl, benzimidazolyl, thiadiazolyl, benzopyranyl, benzothiazolyl, tetrahydrofuryl, tetrahydropyranyl, thienyl (also historically called thiophenyl), benzothienyl, thiamorpholinyl, oxadiazolyl, triazolyl and tetrahydroquinolinyl.

Hydrocarbyloxy refers to groups of from 1 to 20 carbon atoms, preferably 1 to 10 carbon atoms, more preferably 1 to 6 carbon atoms attached to the parent structure through an oxygen. Alkoxy is a subset of hydrocarbyloxy and includes groups of a straight or branched configuration. Examples include methoxy, ethoxy, propoxy, isopropoxy and the like. Lower-alkoxy refers to groups containing one to four carbons.

The term “halogen” means fluorine, chlorine, bromine or iodine. In one embodiment, halogen may be fluorine or chlorine.

Unless otherwise specified, acyl refers to formyl and to groups of 1, 2, 3, 4, 5, 6, 7 and 8 carbon atoms of a straight, branched, cyclic configuration, saturated, unsaturated and aromatic and combinations thereof, attached to the parent structure through a carbonyl functionality. Examples include acetyl, benzoyl, propionyl, isobutyryl and the like. Lower-acyl refers to groups containing one to four carbons. The double bonded oxygen, when referred to as a substituent itself is called “oxo”.

As used herein, the term “optionally substituted” may be used interchangeably with “unsubstituted or substituted”. The term “substituted” refers to the replacement of one or more hydrogen atoms in a specified group with a specified radical. For example, substituted alkyl, aryl, cycloalkyl, heterocyclyl etc. refer to alkyl, aryl, cycloalkyl, or heterocyclyl wherein one or more H atoms in each residue are replaced with halogen, haloalkyl, hydrocarbyl, acyl, alkoxyalkyl, hydroxy lower alkyl, carbonyl, phenyl, heteroaryl, benzenesulfonyl, hydroxy, hydrocarbyloxy, haloalkoxy, oxaalkyl, carboxy, alkoxycarbonyl [—C(═O)O-alkyl], alkoxycarbonylamino [HNC(═O)O-alkyl], aminocarbonyl (also known as carboxamido) [—C(═O)NH2], alkylaminocarbonyl [—C(═O)NH-alkyl], dialkylaminocarbonyl [—C(═O)N(alkyl)2], cyano, acetoxy, nitro, amino, alkylamino, dialkylamino, (alkyl)(aryl)aminoalkyl, alkylaminoalkyl (including cycloalkylaminoalkyl), dialkylaminoalkyl, dialkylaminoalkoxy, heterocyclylalkoxy, mercapto, alkylthio, sulfoxide, sulfone, sulfonylamino, alkylsulfinyl, alkylsulfonyl, acylaminoalkyl, acylaminoalkoxy, acylamino, amidino, aryl, benzyl, heterocyclyl, heterocyclylalkyl, phenoxy, benzyloxy, heteroaryloxy, hydroxyimino, alkoxyimino, oxaalkyl, aminosulfonyl, trityl, amidino, guanidino, ureido, benzyloxyphenyl, and benzyloxy. “Oxo” is also included among the substituents referred to in “optionally substituted”; it will be appreciated by persons of skill in the art that, because oxo is a divalent radical, there are circumstances in which it will not be appropriate as a substituent (e.g. on phenyl). In one embodiment, 1, 2, or 3 hydrogen atoms are replaced with a specified radical. In the case of alkyl and cycloalkyl, more than three hydrogen atoms can be replaced by fluorine; indeed, all available hydrogen atoms could be replaced by fluorine. In particular embodiments, substituents are halogen, halo(C1-C4)hydrocarbyl, halo(C1-C4)hydrocarbyloxy, cyano, thiocyanato, (C1-C4)hydrocarbylsulfinyl, (C1-C4)hydrocarbyl-sulfonyl, aminosulfonyl, nitro, acetyl, and acetamido. Preferred substituents are halogen, (C1-C4)alkyl, (C1-C4)alkoxy, (C1-C4)fluoroalkyl, (C1-C4)fluoroalkoxy, hydroxy, amino, (C1-C4)alkylamino, di(C1-C4)alkylamino, (C1-C4)acylamino, (C1-C4)fluoroalkyl and (C1-C4)fluoroalkoxy.

Substituents Rn are generally defined when introduced and retain that definition throughout the specification and claims.

As used herein, and as would be understood by the person of skill in the art, the recitation of “a compound”—unless expressly further limited—is intended to include salts of that compound. Thus, for example, the recitation “a compound of formula I” as depicted above, which contains a basic amine residue —NR2R3, would include salts —NHR2R3+X wherein X is any counterion. In a particular embodiment, the term “compound of formula I” refers to the compound or a pharmaceutically acceptable salt thereof; this term refers to a pharmaceutically acceptable salt of the compound, even if not explicitly stated. Unless otherwise stated or depicted, structures depicted herein are also meant to include all stereoisomeric (e.g., enantiomeric, diastereomeric, and cis-trans isomeric) forms of the structure; for example, the R and S configurations for each asymmetric center, (Z) and (E) double bond isomers, and (Z) and (E) conformational isomers. Therefore, single stereochemical isomers as well as enantiomeric, diastereomeric, and cis-trans isomeric (or conformational) mixtures of the present compounds are within the scope of the invention. Unless otherwise stated, all tautomeric forms of the compounds of the invention are within the scope of the invention. Additionally, unless otherwise stated, structures depicted herein are also meant to include compounds that differ only in the presence of one or more isotopically enriched atoms. For example, compounds having the present structures except for the replacement of hydrogen by deuterium or tritium, or the replacement of a carbon by a 13C- or 14C-enriched carbon are within the scope of this invention. In addition to therapeutic uses, such compounds are useful, for example, as analytical tools or probes in biological assays.

As used herein, the term “pharmaceutically acceptable salt” refers to those salts which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and lower animals without undue toxicity, irritation, allergic response and the like, and are commensurate with a reasonable benefit/risk ratio. Pharmaceutically acceptable salts are well known in the art. For example, S. M. Berge et al., describe pharmaceutically acceptable salts in detail in J. Pharmaceutical Sciences, 1977, 66, 1-19. Pharmaceutically acceptable salts of the compounds of this invention include those derived from suitable inorganic and organic acids and bases. Examples of pharmaceutically acceptable, nontoxic acid addition salts are salts of an amino group formed with inorganic acids such as hydrochloric acid, hydrobromic acid, phosphoric acid, sulfuric acid and perchloric acid or with organic acids such as acetic acid, oxalic acid, maleic acid, tartaric acid, citric acid, succinic acid or malonic acid or by using other methods used in the art such as ion exchange. Other pharmaceutically acceptable salts include adipate, alginate, ascorbate, aspartate, benzenesulfonate, benzoate, bisulfate, borate, butyrate, camphorate, camphorsulfonate, citrate, cyclopentanepropionate, digluconate, dodecylsulfate, ethanesulfonate, formate, fumarate, glucoheptonate, glycerophosphate, gluconate, hemisulfate, heptanoate, hexanoate, hydroiodide, 2-hydroxy-ethanesulfonate, lactobionate, lactate, laurate, lauryl sulfate, malate, maleate, malonate, methanesulfonate, 2-naphthalenesulfonate, nicotinate, nitrate, oleate, oxalate, palmitate, pamoate, pectinate, persulfate, 3-phenylpropionate, phosphate, pivalate, propionate, stearate, succinate, sulfate, tartrate, thiocyanate, p-toluenesulfonate, undecanoate, valerate salts, and the like. Although pharmaceutically acceptable counter ions will be preferred for preparing pharmaceutical formulations, other anions are quite acceptable as synthetic intermediates. Thus X may be pharmaceutically undesirable anions, such as iodide, oxalate, trifluoromethanesulfonate and the like, when such salts are chemical intermediates.

Unless otherwise specified, the word “includes” (or any variation thereon, e.g., “include”, “including”, etc.) is intended to be open-ended. For example, “A includes 1, 2 and 3” means that A includes but is not limited to 1, 2 and 3.

Unless otherwise specified, the phrase “such as” is intended to be open-ended. For example, “A can be a halogen, such as chlorine or bromine” means that A can be, but is not limited to, chlorine or bromine.

According to another embodiment, the invention provides a composition comprising a compound of this invention (or its pharmaceutically acceptable salt) and a pharmaceutically acceptable carrier, adjuvant, or vehicle. In some embodiments, the amount of compound in compositions of this invention (also referred to herein as “effective amount” or “therapeutically effective amount”) is such that is effective to treat, prevent, and/or manage various neurological and/or psychiatric diseases and disorders and/or symptoms in a subject. In some embodiments, a composition of this invention is formulated for administration to a subject in need of such composition. In some embodiments, a composition of this invention is formulated for oral administration to a subject.

As used herein, the term “subject” to which administration is contemplated includes, but is not limited to, humans (i.e., a male or female of any age group, e.g., a pediatric subject (e.g., infant, child, adolescent) or adult subject (e.g., young adult, middle-aged adult or senior adult)) and/or other primates (e.g., cynomolgus monkeys, rhesus monkeys); mammals, including commercially relevant mammals such as cattle, pigs, horses, sheep, goats, cats, and/or dogs; and/or birds, including commercially relevant birds such as chickens, ducks, geese, quail, and/or turkeys.

In certain embodiments, provided herein is a composition (e.g., a pharmaceutical composition) comprising a compound described herein and a pharmaceutically acceptable excipient or carrier. In some embodiments, provided herein is a method of treating neurological or psychiatric diseases and disorders in a subject in need thereof in a subject, comprising administering an effective amount of a compound or a pharmaceutical composition described herein. Examples of carriers and excipients are well known to those skilled in the art and are described in detail in, e.g., Ansel, Howard C., et al., Ansel's Pharmaceutical Dosage Forms and Drug Delivery Systems. Philadelphia: Lippincott, Williams & Wilkins, 2004; Gennaro, Alfonso R., et al. Remington: The Science and Practice of Pharmacy. Philadelphia: Lippincott, Williams & Wilkins, 2000; and Rowe, Raymond C. Handbook of Pharmaceutical Excipients. Chicago, Pharmaceutical Press, 2005. The formulations may also include one or more buffers, stabilizing agents, surfactants, wetting agents, lubricating agents, emulsifiers, suspending agents, preservatives, antioxidants, opaquing agents, glidants, processing aids, colorants, sweeteners, perfuming agents, flavoring agents, diluents and other known additives to provide an elegant presentation of the drug (i.e., a compound of the present invention or pharmaceutical composition thereof) or aid in the manufacturing of the pharmaceutical product (i.e., medicament).

Compositions of the present invention may be administered orally, parenterally, by inhalation spray, topically, rectally, nasally, buccally, vaginally or via an implanted reservoir. The term “parenteral” as used herein includes subcutaneous, intravenous, intramuscular, intra-articular, intra-synovial, intrasternal, intrathecal, intrahepatic, intralesional and intracranial injection or infusion techniques. Preferably, the compositions are administered orally, intraperitoneally or intravenously. Sterile injectable forms of the compositions of this invention may be aqueous or oleaginous suspension. These suspensions may be formulated according to techniques known in the art using suitable dispersing or wetting agents and suspending agents. The sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally acceptable diluent or solvent, for example as a solution in 1,3-butanediol. Among the acceptable vehicles and solvents that may be employed are water, Ringer's solution and isotonic sodium chloride solution. In addition, sterile, fixed oils are conventionally employed as a solvent or suspending medium. Pharmaceutically acceptable compositions of this invention may be orally administered in any orally acceptable dosage form including capsules, tablets, aqueous suspensions or solutions.

The amount of compounds of the present invention that may be combined with the carrier materials to produce a composition in a single dosage form will vary depending upon a variety of factors, including the host treated and the particular mode of administration. It should also be understood that a specific dosage and treatment regimen for any particular subject will depend upon a variety of factors, including the activity of the specific compound employed, the age, body weight, general health, sex, diet, time of administration, rate of excretion, drug combination, and the judgment of the treating physician and the severity of the particular disease being treated. The amount of a compound of the present invention in the composition will also depend upon the particular compound in the composition.

As used herein, the terms “treatment,” “treat,” and “treating” refer to reversing, alleviating, delaying the onset of, or inhibiting the progress of a disease or disorder, or one or more symptoms thereof, as described herein. In some embodiments, treatment may be administered after one or more symptoms have developed. In other embodiments, treatment may be administered in the absence of symptoms. For example, treatment may be administered to a susceptible individual prior to the onset of symptoms (e.g., in light of a history of symptoms and/or in light of genetic or other susceptibility factors). Treatment may also be continued after symptoms have resolved, for example to prevent or delay their recurrence.

In one embodiment, the invention provides for a method for treating a neurological or psychiatric disease or disorder, wherein the neurological or psychiatric disease or disorder is anxiety or attention deficit hyperactivity disorder.

In one embodiment, provided is a method of treating anxiety or attention deficit hyperactivity disorder in a subject in need thereof, comprising the step of administering to said subject an effective amount of a compound selected from:


or a pharmaceutically acceptable salt thereof.

In one embodiment, provided is a method of treating anxiety in a subject in need thereof, comprising the step of administering to said subject an effective amount of a compound selected from:


or a pharmaceutically acceptable salt thereof.

In one embodiment of the invention, provided is a method of treating attention deficit hyperactivity disorder in a subject in need thereof, comprising the step of administering to said subject an effective amount of a compound selected from:


or a pharmaceutically acceptable salt thereof.

In some embodiments, the invention provides a method for treating a neurological or psychiatric diseases or disorder in a subject, comprising administering to the subject an effective amount of a compound of this invention (or its pharmaceutically acceptable salt), or composition comprising a compound of this invention (or its pharmaceutically acceptable salt). Neurological and/or psychiatric diseases and disorders diseases can exhibit a variety of psychiatric and behavioral symptoms, including apathy, depression, anxiety, cognitive impairment, psychosis, aggression, agitation, poor impulse control and sleep disruptions.

In some embodiments, the neurological or psychiatric diseases or disorder is selected from a psychosis, including schizophrenia (paranoid, disorganized, catatonic or undifferentiated), schizophreniform disorder, schizoaffective disorder, delusional disorder, brief psychotic disorder, shared psychotic disorder, psychotic disorder due to a general medical condition and substance-induced or drug-induced (e.g., phencyclidine, ketamine and other dissociative anesthetics, amphetamine and other psychostimulants and cocaine) psychosispsychotic disorder, psychosis associated with affective disorders, brief reactive psychosis, schizoaffective psychosis, “schizophrenia-spectrum” disorders such as schizoid or schizotypal personality disorders, or illness associated with psychosis (such as major depression, manic depressive (bipolar) disorder, Alzheimer's disease and post-traumatic stress syndrome), including both positive, negative, and cognitive symptoms of schizophrenia and other psychoses; cognitive disorders including dementia (semantic dementia, frontotemporal dementia, dementia with depressive features, persisting, subcortical dementia, dementia with Lewy Bodies, Parkinsonism-ALS Dementia Complex, and dementia associated with Alzheimer's disease, ischemia, multi-infarct dementia, trauma, vascular problems, stroke, HIV disease, Parkinson's disease, Huntington's disease, Down syndrome, Pick's disease, Creutzfeldt-Jacob disease, perinatal hypoxia, or substance abuse), delirium, amnestic disorders or age related cognitive decline; anxiety disorders including acute stress disorder, agoraphobia, generalized anxiety disorder, obsessive-compulsive disorder, panic attack, panic disorder, post-traumatic stress disorder, separation anxiety disorder, social phobia, specific phobia, substance-induced anxiety disorder and anxiety due to a general medical condition; substance-related disorders and addictive behaviors (including substance-induced delirium, persisting dementia, persisting amnestic disorder, psychotic disorder or anxiety disorder; tolerance, dependence or withdrawal from substances including alcohol, amphetamines, cannabis, cocaine, hallucinogens, inhalants, nicotine, opioids, phencyclidine, sedatives, hypnotics or anxiolytics); obesity, bulimia nervosa, pica and compulsive eating disorders; bipolar disorders, mood disorders including depressive disorders; depression including unipolar depression, seasonal depression and post-partum depression, atypical depression, catatonic depression, elderly depression, endogenous depression, melancholic depression, perinatal depression, situational depression, chronic depression, premenstrual syndrome (PMS) and premenstrual dysphoric disorder (PDD), mood disorders due to a general medical condition, and substance-induced mood disorders; learning disorders, pervasive developmental disorder including autistic disorder, attention disorders including attention-deficit hyperactivity disorder (ADHD) and conduct disorder; disorders such as autism, depression, benign forgetfulness, childhood learning disorders, specific learning disorders, intellectual development disorders, and closed head injury; movement disorders; epilepsy; urinary incontinence; neuronal damage including ocular damage, retinopathy or macular degeneration of the eye, tinnitus, hearing impairment and loss, and brain edema; emesis; and sleep disorders including insomnia and narcolepsy.

In some embodiments, the neurological or psychiatric disease or disorder is Alzheimer's disease, Parkinson's disease, depression, cognitive impairment, stroke, schizophrenia, Down syndrome, or Fetal Alcohol Syndrome. In some embodiments, the neurological or psychiatric disease or disorder is Alzheimer's disease. In some embodiments, the neurological or psychiatric disease or disorder is Parkinson's disease. In some embodiments, the neurological or psychiatric disease or disorder is depression. In some embodiments, the neurological or psychiatric disease disorder is cognitive impairment. In some embodiments, the cognitive impairment is cognitive dysfunction associated with depression, for example, major depressive disorder. In some embodiments, the neurological or psychiatric disorder is stroke. In some embodiments, the neurological or psychiatric disease or disorder is schizophrenia. In some embodiments, the neurological or psychiatric disease or disorder is Down syndrome. In some embodiments, the neurological or psychiatric disorder is Fetal Alcohol Syndrome.

In some embodiments, the neurological or psychiatric disease or disorder is bipolar disease. Bipolar disorder is a serious psychiatric disorder that has a prevalence of approximately 2% of the population, and affects both genders alike. It is a relapsing-remitting condition characterized by cycling between elevated (i.e., manic) and depressed moods, which distinguishes it from other disorders such as major depressive disorder and schizophrenia. Bipolar I is defined by the occurrence of a full manic episode, although most individuals experience significant depression. Symptoms of mania include elevated or irritable mood, hyperactivity, grandiosity, decreased need for sleep, racing thoughts and in some cases, psychosis. The depressive episodes are characterized by anhedonia, sad mood, hopelessness, poor self-esteem, diminished concentration and lethargy. Bipolar II is defined as the occurrence of a major depressive episode and hypomanic (less severe mania) episode although subjects spend considerable more time in the depressive state. Other related conditions include cyclothymic disorder.

In some embodiments, the neurological or psychiatric disease or disorder is schizophrenia. Schizophrenia is a disorder of unknown origin, which usually appears for the first time in early adulthood and is marked by characteristics such as psychotic symptoms, phasic progression and development, and/or deterioration in social behavior and professional capability. Characteristic psychotic symptoms are disorders of thought content (e.g., multiple, fragmentary, incoherent, implausible or simply delusional contents, or ideas of persecution) and of mentality (e.g., loss of association, flight of imagination, incoherence up to incomprehensibility), as well as disorders of perceptibility (e.g., hallucinations), emotions (e.g., superficial or inadequate emotions), self-perceptions, intentions, impulses, and/or inter-human relationships, and psychomotoric disorders (e.g., catatonia). Other symptoms are also associated with this disorder. Schizophrenia is classified into subgroups: the paranoid type, characterized by delusions and hallucinations and absence of thought disorder, disorganized behavior, and affective flattening; the disorganized type, also named “hebephrenic schizophrenia,” in which thought disorder and flat affect are present together; the cataconic type, in which prominent psychomotor disturbances are evident, and symptoms may include catatonic stupor and waxy flexibility; and the undifferentiated type, in which psychotic symptoms are present but the criteria for paranoid, disorganized, or catatonic types have not been met. The symptoms of schizophrenia normally manifest themselves in three broad categories: positive, negative and cognitive symptoms. Positive symptoms are those which represent an “excess” of normal experiences, such as hallucinations and delusions. Negative symptoms are those where the subject suffers from a lack of normal experiences, such as anhedonia and lack of social interaction. The cognitive symptoms relate to cognitive impairment in schizophrenics, such as lack of sustained attention and deficits in decision making.

In some embodiments, the neurological or psychiatric disease or disorder is anxiety disorder. Anxiety disorders are characterized by fear, worry, and uneasiness, usually generalized and unfocused as an overreaction to a situation. Anxiety disorders differ in the situations or types of objects that induce fear, anxiety, or avoidance behavior, and the associated cognitive ideation. Anxiety differs from fear in that anxiety is an emotional response to a perceived future threat while fear is associated with a perceived or real immediate threat. They also differ in the content of the associated thoughts or beliefs. Examples of anxiety disorders include separation anxiety disorder, selective mutism, specific phobia, social anxiety disorder (social phobia), panic disorder, panic attack specifier, agoraphobia, generalized anxiety disorder, substance/medication-induced anxiety disorder, anxiety disorder due to another medical condition, illness anxiety disorder, social (pragmatic) communication disorder, other specified anxiety disorder, and unspecified anxiety disorder; stressor-related disorders, including reactive attachment disorder, disinhibited social engagement disorder, posttraumatic stress disorder (PTSD), acute stress disorder, and adjustment disorders.

Cognitive impairment includes a decline in cognitive functions or cognitive domains, e.g., working memory, attention and vigilance, verbal learning and memory, visual learning and memory, reasoning and problem solving (e.g., executive function, speed of processing and/or social cognition). In particular, cognitive impairment may indicate deficits in attention, disorganized thinking, slow thinking, difficulty in understanding, poor concentration, impairment of problem solving, poor memory, difficulties in expressing thoughts, and/or difficulties in integrating thoughts, feelings and behavior, or difficulties in extinction of irrelevant thoughts.

In some embodiments, the neurological or psychiatric disease or disorder involves a deficit in cognition (cognitive domains as defined by the DSM-5 are: complex attention, executive function, learning and memory, language, perceptual-motor, social cognition). In some embodiments, the neurological or psychiatric disorder is associated with a deficit in dopamine signaling. In some embodiments, the neurological or psychiatric disorder is associated with basal ganglia dysfunction. In some embodiments, the neurological or psychiatric disorder is associated with dysregulated locomotor activity. In some embodiments, the neurological or psychiatric disorder is associated with impairment of prefrontal cortex functioning.

In some embodiments, the present invention provides a method of treating one or more symptoms of a neurological and/or psychiatric disease or disorder provided herein. Such diseases and disorders include mood disorders, including bipolar I disorder, bipolar II disorder, bipolar depression, mania, cyclothymic disorder, sub stance/medication-induced bipolar and related disorders, bipolar and related disorder due to another medical condition, other specified bipolar and related disorder, and unspecified bipolar and related disorders; psychotic disorders, including schizophrenia, schizophrenia spectrum disorder, acute schizophrenia, chronic schizophrenia, NOS schizophrenia, schizoid personality disorder, schizotypal personality disorder, delusional disorder, psychosis, psychotic disorder, brief psychotic disorder, shared psychotic disorder, psychotic disorder due to a general medical condition, drug-induced psychosis (e.g., cocaine, alcohol, amphetamine), schizoaffective disorder, agitation, aggression, delirium, catalepsy, catatonia, dissociative identity disorder, paranoid personality disorder, psychotic depression, Schizotypical Personality Disorder, Childhood Disintegrative Disorder (Heller's Syndrome), Disintegrative Psychosis, Dissociative Amnesia, Somatic Symptom Disorder, Parkinson's psychosis, excitative psychosis, Tourette's syndrome, and organic or NOS psychosis; depressive disorders, including disruptive mood dysregulation disorder, major depressive disorder (MDD) (including major depressive episode), dysthymia, persistent depressive disorder (dysthymia), treatment resistant depression, premenstrual dysphoric disorder, substance/medication-induced depressive disorder, depressive disorder due to another medical condition, other specified depressive disorder, and unspecified depressive disorder; anxiety disorders; and other disorders including substance abuse or dependency (e.g., nicotine, alcohol, cocaine), addiction, internet gaming disorder, eating disorders, behavior disorder, seizure, vertigo, epilepsy, agitation, aggression, neurodegenerative disease, Alzheimer's disease, Parkinson's disease, dyskinesias, Huntington's disease, dementia, premenstrual dysphoria, attention deficit disorder (ADD) and attention deficit hyperactivity disorder (ADHD)), hyperkinetic syndrome, autism, autism spectrum disorder, obsessive-compulsive disorder, pain, fibromyalgia, migraine, cognitive impairment, movement disorder, restless leg syndrome (RLS), multiple sclerosis, Primary Progressive Multiple Sclerosis, multiple sclerosis, sleep disorder, sleep apnea, narcolepsy, excessive daytime sleepiness, jet lag, drowsy side effect of medications, insomnia, sexual dysfunction, hypertension, emesis, Lesche-Nyhane disease, Wilson's disease, Rett syndrome, and Huntington's chorea. In some embodiments, the neurological and/or psychiatric disorders include agitation and aggression.

In some embodiments, the agitation and aggression are associated with Alzheimer's disease, Parkinson's disease, and/or autism.

In some embodiments, the neurological and/or psychiatric diseases or disorders are obsessive-compulsive disorder and related disorders (e.g., body dysmorphic disorder, hoarding disorder, trichotillomania, excoriation disorder).

In some embodiments, the neurological and/or psychiatric diseases or disorders are disruptive, impulse-control, and conduct disorders including oppositional defiant disorder, intermittent explosive disorder, conduct disorder, antisocial personality disorder, pyromania, kleptomania, other specified disruptive, impulse-control, and conduct disorder, unspecified disruptive, impulse-control, and conduct disorder.

Depressive disorders include major depressive disorder and dysthymia, and are associated with depressed mood (sadness), poor concentration, insomnia, fatigue, appetite disturbances, excessive guilt and thoughts of suicide.

In some embodiments, the present invention provides a method of treating one or more symptoms including depression (e.g., major depressive disorder or dysthymia); bipolar disorder, seasonal affective disorder; cognitive deficit; sleep related disorder (e.g., sleep apnea, insomnia, narcolepsy, cataplexy) including those sleep disorders which are produced by psychiatric conditions; chronic fatigue syndrome; anxieties (e.g., general anxiety disorder, social anxiety disorder, panic disorder); obsessive compulsive disorder; post-menopausal vasomotor symptoms (e.g., hot flashes, night sweats); neurodegenerative disease (e.g., Parkinson's disease, Alzheimer's disease, amyotrophic lateral sclerosis, primary lateral sclerosis, progressive muscular atrophy, progressive bulbar (atrophy) palsy, pseudobulbar palsy spinal muscular atrophy diseases (e.g., SMA type I, also called Werdnig-Hoffmann disease, SMA type II, SMA type III, also called Kugelberg-Welander disease, and Kennedy Disease, also called progressive spinobulbar muscular atrophy), Hallervorden-Spatz disease, Seitelberger disease (Infantile Neuroaxonal Dystrophy), adrenoleukodystrophy, Alexander Disease, autosomal dominant cerebellar ataxia (ADCA), pure autonomic failure (Bradbury-Eggleston Syndrome), CADASIL Syndrome, and neuronal ceroids lipofuscinose disorders such as Batten Disease (Spielmeyer-Vogt-Sjögren)); manic disorder; dysthymic disorder; and obesity.

In some embodiments, a depressive disorder is associated with acute suicidality or suicide ideation. The United States Food and Drug Administration has adopted a “black box” label warning indicating that antidepressants may increase the risk of suicidal thinking and behavior in some children, adolescents and young adults (up to age 24) with a depressive disorder such as MDD. In some embodiments, a provided compound does not increase the risk of suicidal thinking and/or behavior in children, adolescents and/or young adults with a depressive disorder, e.g., with MDD. In some embodiments, the present invention provides a method of treating one or more symptoms of a depressive disorder (e.g., MDD) in children, adolescents and/or young adults without increasing the risk of suicidal thinking and/or behavior.

In some embodiments, the present invention provides a method of treating one or more symptoms including senile dementia, Early Onset Alzheimer's Disease, Alzheimer's type dementia, cognition, memory loss, amnesia/amnestic syndrome, disturbances of consciousness, coma, lowering of attention, speech disorder, agnosia, aphasia, apraxia, Mild Cognitive Impairment (MCI), benign forgetfulness, mild neurocognitive disorder, major neurocognitive disorder, neurocognitive disorder due to disease (e.g., Huntington's Disease, Parkinson's disease, Prion Disease, Traumatic Brain Injury, HIV or AIDS), Binswanger's Disease (subcortical leukoencephalopathy), and Capgras Syndrome.

In some embodiments, the present invention provides a method of treating one or more symptoms of pain, e.g., neuropathic pain, sensitization accompanying neuropathic pain, or inflammatory pain. In some embodiments, the pain is neuropathic pain, including post herpetic (or post-shingles) neuralgia, reflex sympathetic dystrophy/causalgia or nerve trauma, phantom limb pain, carpal tunnel syndrome, and peripheral neuropathy (such as diabetic neuropathy or neuropathy arising from chronic alcohol use). In some embodiments, the pain is acute pain, nociceptive pain, arthritis pain, rheumatoid arthritis, osteoarthritis, joint pain, muscoskeletal pain, back pain, dorsalgia, bulging disc, hip pain, visceral pain, headache, tension headache, acute tension headache, chronic tension headache, chronic cluster headache, common migraine, classic migraine, cluster headache, mixed headache, post-traumatic headache, eye strain headache, Short-lasting Unilateral Neuralgiform (SUNCT) headache, SUNCT Syndrome, herpes zoster, acute herpes zoster, shingles, postherpetic neuralgia (shingles), causalgia, central pain, central pain syndrome, chronic back pain, neuralgia, neuropathic pain syndrome, neuropathy, diabetic neuropathy, diabetes-related neuropathy, diabetes-related nerve pain, fibrositis, peripheral neuropathy caused by chemotherapy, peripheral nerve disease, peripheral neuropathy, nerve pain, nerve trauma, sensitization accompanying neuropathic pain, complex regional pain syndrome, compression neuropathy, craniofacial pain, chronic joint pain, chronic knee pain, chronic pain syndrome, cancer pain, trigeminal neuralgia, tic doloreaux, reflex sympathetic causalgia, painful peripheral neuropathy, spinal nerve injury, arachnoiditis, spinal pain, Bernhardt-Roth Syndrome (meralgia parasthetica), carpal tunnel syndrome, cerebrospinal fluid syndrome, Charcot-Marie-tooth disease, hereditary motor and sensory neuropathy, peroneal muscular atrophy, cluster-tic syndrome, coccygeal pain syndromes, compartment syndrome, degenerative disc disease, failed back surgery syndrome, genito-pelvic pain/penetration disorder, gout, inflammatory pain, lumbar radiculopathy, neuroma (painful scar), pain associated with multiple sclerosis, pelvic floor disorders, phantom limb pain, piriformis syndrome, psychogenic pain, radicular pain syndrome, Raeder's syndrome, referred pain, reflex sympathetic dystrophy syndrome, sciatica, sciatica pain, scoliosis, slipped disc, somatic pain, spinal stenosis, stiff-person syndrome/stiff-man syndrome, stump pain, sympathetically maintained pain, tolosa-hunt syndrome, whiplash, or pain associated with Lyme disease.

In some embodiments, the present invention provides a method of treating one or more symptoms including obesity; migraine or migraine headache; and sexual dysfunction, in men or women, including without limitation sexual dysfunction caused by psychological and/or physiological factors, erectile dysfunction, premature ejaculation, vaginal dryness, lack of sexual excitement, inability to obtain orgasm, and psycho-sexual dysfunction, including without limitation, inhibited sexual desire, inhibited sexual excitement, inhibited female orgasm, inhibited male orgasm, functional dyspareunia, functional vaginismus, and atypical psychosexual dysfunction.

In some embodiments, the present invention provides a method of suppressing rapid eye movement (REM) during both sleep and daytime equivalent.

In some embodiments, the present invention provides a method of suppressing or eliminating pathological or excessive REM during the night or daytime equivalent.

In some embodiments, the present invention provides a method of treating one or more symptoms including cataplexy (sudden involuntary transient bouts of muscle weakness or paralysis while awake); nighttime sleep disturbance/sleep fragmentation associated with narcolepsy or other conditions; sleep paralysis associated with narcolepsy or other conditions; hypnagogic and hypnapompic hallucinations associated with narcolepsy or other conditions; and excessive daytime sleepiness associated with narcolepsy, sleep apnea or shift work disorder and other medical conditions such as cancer, chronic fatigue syndrome and fibromyalgia.

In some embodiments, the present invention provides a method of treating one or more symptoms of movement disorders, including akinesias, akinetic-rigid syndromes, dyskinesias and dystonias. Examples of akinesias and akinetic-rigid syndromes include Parkinson's disease, drug-induced Parkinsonism, postencephalitic Parkinsonism, secondary Parkinsonism, Parkinson plus syndromes, atypical Parkinsonism, idiopathic Parkinsonism, progressive supranuclear palsy, multiple system atrophy, corticobasal degeneration, Parkinsonism-ALS dementia complex and basal ganglia calcification, medication-induced Parkinsonism (such as neuroleptic-induced parkinsonism, neuroleptic malignant syndrome, neuroleptic-induced acute dystonia, neuroleptic-induced acute akathisia, neuroleptic-induced tardive dyskinesia and medication-induced postural tremor), Gilles de la Tourette's syndrome, epilepsy, muscular spasms and disorders associated with muscular spasticity or weakness including tremors. Examples of dyskinesias include drug (e.g. L-DOPA) induced dyskinesia tremor (such as rest tremor, postural tremor, intention tremor), chorea (such as Sydenham's chorea, Huntington's disease, benign hereditary chorea, neuroacanthocytosis, symptomatic chorea, drug-induced chorea and hemiballism), myoclonus (including generalised myoclonus and focal myoclonus), tics (including simple tics, complex tics and symptomatic tics). Examples of dystonias include generalised dystonia, iodiopathic dystonia, drug-induced dystonia, symptomatic dystonia, paroxymal dystonia, focal dystonia, blepharospasm, oromandibular dystonia, spasmodic dysphonia, spasmodic torticollis, axial dystonia, dystonic writer's cramp and hemiplegic dystonia. Other examples of movement disorders include stereotypic movement disorder, persistent (chronic) motor disorder, medication-Induced movement disorder, psychogenic movement disorders, sub stance/medication-Induced movement disorder, extrapyramidal movement disorders, hyperkinetic movement disorders, hypokinetic movement disorders, alternating hemiplegia, Angelman syndrome, Hallervorden-Spatz Disease, ataxia, dentate cerebellar ataxia, ataxia telangiectasia (Louis-Bar syndrome), Friedreich's Ataxia, hereditary spinal ataxia, hereditary spinal sclerosis, Machado-Joseph Disease, spinocerebellar ataxia, progressive myoclonic ataxia, athetosis, ballismus, blepharospasm (eye twitching), cerebral palsy, tardive dystonia, tardive dyskinesia, idiopathic torsion dystonia, torsion dystonia, focal dystonia, idiopathic familial dystonia, Idiopathic nonfamilial dystonia, cervical dystonia (spasmodic torticollis), primary dystonia, orofacial dystonia, developmental coordination disorder, bulbospinal muscular atrophy (Kennedy's Disease), Shy-Drager Syndrome, and Stiff-Person (Stiff-Man) Syndrome.

In some embodiments, the present invention provides a method of treating one or more symptoms of epilepsy and/or seizures, including abdominal epilepsy, absence seizure, acquired epilepsy, acquired epileptiform aphasia, Aicardi syndrome, Alpers' disease, Alpers-Huttenlocher syndrome, Angelman syndrome, benign focal epilepsy, benign focal epilepsy of childhood, benign intracranial hypertension, benign rolandic epilepsy (BRE), CDKL5 disorder, childhood absence epilepsy, dentate cerebellar ataxia, Doose syndrome, Dravet syndrome, dyscognitive focal seizure, epilepsy with grand mal seizures, epilepsy with myoclonic-absences, epileptic hemiplegia, febrile seizures, focal seizure, frontal lobe epilepsy, generalized tonic-clonic seizures, genetic epilepsy, Glutl deficiency syndrome, hypothalmic hamartoma, idiopathic epilepsy, idiopathic generalized epilepsy, idopathic localization-related epilepsies, idopathic partial epilepsy, idopathic seizure, juvenile absence epilepsy, juvenile myoclonic epilepsy, Lafora disease, Lafora progressive myoclonus epilepsy, Landau-Kleffner syndrome, Lassueur-Graham-Little syndrome, Lennox syndrome, Lennox-Gastaut syndrome, medically refractory epilepsy, mesial-temporal lobe sclerosis, myoclonic seizure, neonatal epilepsy, occipital lobe epilepsy, Ohtahara syndrome, Panayiotopoulos syndrome, parietal lobe epilepsy, PCDH19 epilepsy, photosensitive epilepsy, progressive myoclonic epilepsies, Rasmussen's encephalitis, Rasmussen's syndrome, refractory epilepsy, seizure disorder, status epilepticus, Sturge-Weber syndrome, symptomatic generalized epilepsy, symptomatic parital epilepsy, TBCK-related ID syndrome, temporal lobe epilepsy, temporal lobe seizures, tonic-clonic seizure, West syndrome, tremor, cerebellar tremor, cerebellar outflow tremor, intention tremor, essential tremor, benign essential tremor, Parkinsonian tremor, and medication-induced postural tremor.

In some embodiments, the present invention provides a method of treating a neurological and/or psychiatric disease or disorder described herein, comprising administering a compound of the invention in conjunction with one or more pharmaceutical agents. Suitable pharmaceutical agents that may be used in combination with the compounds of the present invention include anti-Parkinson's drugs, anti-Alzheimer's drugs, anti-depressants, anti-psychotics, anti-ischemics, CNS depressants, anti-cholinergics, nootropics, epilepsy medication, attention (e.g., ADD/ADHD) medications, sleep-promoting medications, wakefulness-promoting medications, and pain medications. In some embodiments, suitable pharmaceutical agents are anxiolytics.

Suitable anti-Parkinson's drugs include dopamine replacement therapy (e.g. L-DOPA, carbidopa, COMT inhibitors such as entacapone or tolcapone), dopamine agonists (e.g. D1 agonists, D2 agonists, mixed D1/D2 agonists, bromocriptine, pergolide, cabergoline, ropinirole, pramipexole, piribedil, or apomorphine in combination with domperidone), histamine H2 antagonists, monoamine oxidase inhibitors (such as selegiline, rasagiline, safinamideand tranylcypromine), certain atypical antipsychotics such as pimavanserin (a non-dopaminergic atypical antipsychotic and inverse agonist of the serotonin 5-HT2A receptor), and amantadine.

In some embodiments, compounds of the invention can be used in combination with levodopa (with or without a selective extracerebral decarboxylase inhibitor such as carbidopa or benserazide), anticholinergics such as biperiden (optionally as its hydrochloride or lactate salt) and trihexyphenidyl(benzhexyl)hydrochloride, COMT inhibitors such as entacapone or tolcapone, MAO A/B inhibitors, antioxidants, A2a adenosine receptor antagonists, cholinergic agonists, NMDA receptor antagonists, serotonin receptor antagonists and dopamine receptor agonists such as alentemol, bromocriptine, fenoldopam, lisuride, naxagolide, pergolide and pramipexole. It will be appreciated that the dopamine agonist may be in the form of a pharmaceutically acceptable salt, for example, alentemol hydrobromide, bromocriptine mesylate, fenoldopam mesylate, naxagolide hydrochloride and pergolide mesylate. Lisuride and pramipexole are commonly used in a non-salt form.

Suitable anti-Alzheimer's drugs include beta-secretase inhibitors, gamma-secretase inhibitors, cholinesterase inhibitors such as donepezil, galantamine or rivastigmine, HMG-CoA reductase inhibitors, NSAID's including ibuprofen, vitamin E, and anti-amyloid antibodies. In some embodiments, an anti-Alzheimer's drug is memantine.

Suitable anti-depressants and anti-anxiety agents include norepinephrine reuptake inhibitors (including tertiary amine tricyclics and secondary amine tricyclics), selective serotonin reuptake inhibitors (SSRIs), monoamine oxidase inhibitors (MAOIs), reversible inhibitors of monoamine oxidase (RIMAs), serotonin and noradrenaline reuptake inhibitors (SNRIs), corticotropin releasing factor (CRF) antagonists, α-adrenoreceptor antagonists, neurokinin-1 receptor antagonists, atypical anti-depressants, benzodiazepines, 5-HT1A agonists or antagonists, especially 5-HT1A partial agonists, and corticotropin releasing factor (CRF) antagonists.

Specific suitable anti-depressant and anti-anxiety agents include amitriptyline, clomipramine, doxepin, imipramine and trimipramine; amoxapine, desipramine, citalopram, escitalopram, maprotiline, nortriptyline and protriptyline; fluoxetine, fluvoxamine, paroxetine and sertraline; isocarboxazid, phenelzine, tranylcypromine and selegiline; moclobemide: venlafaxine; desvenlafaxine, duloxetine; aprepitant; bupropion, vilazodone, mirtazapine, lithium, nefazodone, trazodone and viloxazine; alprazolam, chlordiazepoxide, clonazepam, chlorazepate, diazepam, halazepam, lorazepam, oxazepam and prazepam; buspirone, flesinoxan, gepirone and ipsapirone, reboxetine, vortioxetine, clorazepate, and pharmaceutically acceptable salts thereof. In some embodiments, suitable anti-depressant and anti-anxiety agents are tianeptine, or pharmaceutically acceptable salts thereof.

Suitable anti-psychotic and mood stabilizer agents include D2 antagonists, 5HT2A antagonists, atypical antipsychotics, lithium, and anticonvulsants.

Specific suitable anti-psychotic and mood stabilizer agents include chlorpromazine, fluphenazine, haloperidol, amisulpride, perphenazine, thioridazine, trifluoperazine, aripiprazole, asenapine, clozapine, olanzapine, paliperidone, quetiapine, risperidone, ziprasidone, lurasidone, flupentixol, levomepromazine, pericyazine, perphenazine, pimozide, prochlorperazine, zuclopenthixol, olanzapine and fluoxetine, lithium, carbamazepine, lamotrigine, valproic acid, iloperidone, thiothixene and pharmaceutically acceptable salts thereof.

Suitable epilepsy medications include levetiracetam, oxcarbazepine, clobazam, retigabine, zonisamide, felbamate, esclicarbazepine acetate, lacosamide, carbamazepine, tiagabine, methsuximide, progabide, valproic acid, lamotrigine, brivaracetam, rufinamide, topiramate and perampanel.

Suitable attention medications include methyl phenidate, atomoxetine, guanfacine, D-amphetamine, lisdexamphetamine, methylamphetamine, and clonidine.

Suitable sleep-promoting medications include ramelteon, triazolam, zopiclone, eszopiclone, zolpidem, temazepam, and trazodone.

Suitable wakefulness-promoting medications include Modafinil, D-Amphetamine, caffeine, and armodafinil.

Suitable pain medications include dextromethorphan, tapentadol, buprenorphine, codeine, fentanyl, hydrocodone, hydromorphone, morphine, naloxegol, oxycodone, tramadol, gabapentil, difluprednate, pregabalin, acetyl salicyclic acid, bromfenac, diclofenac, diflunisal, indomethacin, ketorolac, meoxican, and naproxen.

In some embodiments, compounds of the invention may be used in combination with other therapies. Suitable therapies include psychotherapy, cognitive behavioral therapy, electroconvulsive therapy, transcranial magnetic stimulation, vagus nerve stimulation, and deep-brain stimulation.

The exact amount required will vary from subject to subject, depending on the species, age, and general condition of the subject, the severity of the condition, the particular agent, its mode of administration, and the like. The compounds of the invention are preferably formulated in dosage unit form for ease of administration and uniformity of dosage. The expression “dosage unit form” as used herein refers to a physically discrete unit of agent appropriate for the subject to be treated. It will be understood, however, that the total daily usage of the compounds and compositions of the present invention will be decided by the attending physician within the scope of sound medical judgment.

The pharmaceutically acceptable compositions of this invention can be administered to humans and other animals orally, rectally, parenterally, intracisternally, intravaginally, intraperitoneally, topically (as by powders, ointments, or drops), bucally, as an oral or nasal spray, or the like, depending on the severity of the disease or disorder being treated. In some embodiments, the compounds of the invention may be administered orally or parenterally at dosage levels of about 0.01 mg/kg to about 50 mg/kg and preferably from about 1 mg/kg to about 25 mg/kg, of subject body weight per day, one or more times a day, to obtain the desired therapeutic effect.

In some embodiments, a combination of two or more therapeutic agents may be administered together with the compounds of the invention. In some embodiments, a combination of three or more therapeutic agents may be administered with the compounds of the invention.

Other examples of agents the compounds of this invention may also be combined with include: vitamins and nutritional supplements, antiemetics (e.g. 5-HT3 receptor antagonists, dopamine antagonists, NK1 receptor antagonists, histamine receptor antagonists, cannabinoids, benzodiazepines, or anticholinergics), agents for treating Multiple Sclerosis (MS) such as beta interferon (e.g., Avonex® and Rebif®, dalfampridine, alemtuzumab), Copaxone®, and mitoxantrone; treatments for Huntington's disease such as tetrabenazine; treatments for asthma such as albuterol and Singulair®; anti-inflammatory agents such as corticosteroids, TNF blockers, IL-1 RA, azathioprine, and sulfasalazine; immunomodulatory and immunosuppressive agents such as cyclosporin, tacrolimus, rapamycin, mycophenolate mofetil, interferons, corticosteroids, cyclophosphamide, azathioprine, and sulfasalazine; neurotrophic factors such as acetylcholinesterase inhibitors, MAO inhibitors, interferons, anti-convulsants, ion channel blockers, riluzole, agents for treating cardiovascular disease such as beta-blockers, ACE inhibitors, diuretics, nitrates, calcium channel blockers, and statins, fibrates, cholesterol absorption inhibitors, bile acid sequestrants, and niacin; agents for treating liver disease such as corticosteroids, cholestyramine, interferons, and anti-viral agents; agents for treating blood disorders such as corticosteroids, anti-leukemic agents, and growth factors; agents for treating immunodeficiency disorders such as gamma globulin; and anti-diabetic agents such as biguanides (metformin, phenformin, buformin), thiazolidinediones (rosiglitazone, pioglitazone, troglitazone), sulfonylureas (tolbutamide, acetohexamide, tolazamide, chlorpropamide, glipizide, glyburide, glimepiride, gliclazide), meglitinides (repaglinide, nateglinide), alpha-glucosidase inhibitors (miglitol, acarbose), incretin mimetics (exenatide, liraglutide, taspoglutide), gastric inhibitory peptide analogs, DPP-4 inhibitors (vildagliptin, sitagliptin, saxagliptin, linagliptin, alogliptin), amylin analogs (pramlintide), and insulin and insulin analogs.

In some embodiments, a compound of the present invention, or a pharmaceutically acceptable salt thereof, is administered in combination with an antisense agent, a monoclonal or polyclonal antibody, or a siRNA therapeutic.

Those additional agents may be administered separately from an inventive compound-containing composition, as part of a multiple dosage regimen. Alternatively, those agents may be part of a single dosage form, mixed together with a compound of this invention in a single composition. If administered as part of a multiple dosage regime, the two active agents may be submitted simultaneously, sequentially or within a period of time from one another, normally within five hours from one another.

As used herein, the term “combination,” “combined,” and related terms refers to the simultaneous or sequential administration of therapeutic agents in accordance with this invention. For example, a compound of the present invention may be administered with another therapeutic agent simultaneously or sequentially in separate unit dosage forms or together in a single unit dosage form. Accordingly, the present invention provides a single unit dosage form comprising a compound of formula I, or a pharmaceutically acceptable salt thereof, an additional therapeutic agent, and a pharmaceutically acceptable carrier, adjuvant, or vehicle.

The amount of both, an inventive compound and additional therapeutic agent (in those compositions which comprise an additional therapeutic agent as described above) that may be combined with the carrier materials to produce a single dosage form will vary depending upon the host treated and the particular mode of administration. Preferably, compositions of this invention should be formulated so that a dosage of between 0.01-100 mg/kg body weight/day of an inventive can be administered.

In those compositions which comprise an additional therapeutic agent, that additional therapeutic agent and the compound of this invention may act synergistically. Therefore, the amount of additional therapeutic agent in such compositions will be less than that required in a monotherapy utilizing only that therapeutic agent. In such compositions a dosage of between 0.01-100 mg/kg body weight/day of the additional therapeutic agent can be administered.

The amount of additional therapeutic agent present in the compositions of this invention will be no more than the amount that would normally be administered in a composition comprising that therapeutic agent as the only active agent. Preferably the amount of additional therapeutic agent in the presently disclosed compositions will range from about 50% to 100% of the amount normally present in a composition comprising that agent as the only therapeutically active agent.

In some embodiments, the present invention provides a medicament comprising at least one compound of formula I, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier, adjuvant, or vehicle.

In some embodiments, the present invention provides the use of a compound of formula I, or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for the treatment of a neurological and/or psychiatric disease or disorder.

EXAMPLES

As depicted in the Examples below, in some embodiments, compounds are prepared according to the following procedures. It will be appreciated that, although the general methods depict the synthesis of certain compounds of the present invention, the following methods, and other methods known to persons skilled in the art, can be applied to all compounds and subclasses and species of each of these, as described herein.

Example 1. 6-Chloroisochroman-4-amine hydrochloride

Step 1.

Methyl 2-hydroxyacetate (1.72 g, 19.2 mmol) was dissolved in DMF (50 mL) and treated with sodium hydride (460 mg, 19.2 mmol, 95%). The reaction mixture was stirred at room temperature for 30 min, and then cooled in an ice bath. A solution of 2.53 g (9.60 mmol) of methyl 2-(bromomethyl)-5-chlorobenzoate (Thorarensen, A., et al WO2004018414) in DMF (5 mL) was added, and the reaction mixture was stirred at 0° C. for 10 min. Excess hydride was carefully quenched by the dropwise addition of saturated aqueous NH4Cl (20 mL). The mixture was diluted with water (50 mL) and extracted with EtOAc (4×60 mL). The combined organics were washed with brine (2×25 mL), anhydrous Na2SO4, filtered, and concentrated under reduced pressure. Purification by flash column chromatography (SiO2, 5% EtOAc/hexanes) gave methyl 5-chloro-2-((2-methoxy-2-oxoethoxy)methyl)benzoate 1.1 (1.82 g, 70%) as a white solid. MS (ESI): m/z 273 [M+H].

Step 2.

A solution of compound 1.1 (1.83 g, 6.71 mmol) in EtOH (10 mL) was treated with a solution of NaOH (1.2 g, 30.1 mmol) in water (7 mL), and stirred at room temperature for 1 h. The mixture was acidified to pH=3-4 by the addition of 1M aqueous HCl. The aqueous phase was extracted with DCM (4×50 mL). The combined organics were washed with brine (2×20 mL), dried over anhydrous Na2SO4, filtered, and concentrated under reduced pressure to provide 2-((carboxymethoxy)methyl)-5-chlorobenzoic acid 1.2 (1.59 g, 97%) as a white solid. LC-MS (ESI): m/z 243 [M−H].

Step 3.

Potassium acetate (2.56 g, 26.1 mmol) was added to a solution of compound 1.2 (1.6 g, 6.54 mmol) in Ac2O (30 mL). The reaction mixture was heated to 140° C. and stirred for 2 h. Ice water (100 mL) and Et2O (100 mL) was added to the reaction vessel and the resulting biphasic mixture was transferred to a separatory funnel. The layers were separated and the aqueous phase was extracted with Et2O (5×50 mL). The combined organics were washed with brine (2×20 mL), dried over anhydrous Na2SO4, filtered, and concentrated under reduced pressure to provide acetic 6-chloro-4-oxoisochroman-3-carboxylic anhydride 1.3 (1.02 g, 15%) as brown oil, which was used without further purification.

Step 4.

A solution of compound 1.3 (1.02 g, 3.83 mmol) in EtOH (30 mL) was treated with a solution of NaOH (306 mg, 7.66 mmol) in water (10 mL). The reaction mixture was stirred at ambient temperature for 15 min and then extracted with EtOAc (4×10 mL). The combined organics were washed with brine, dried over anhydrous Na2SO4, filtered, and concentrated under reduced pressure. Purification by flash column chromatography (SiO2, 5% EtOAc/hexanes) afforded 6-chloroisochroman-4-one 1.4 (0.505 g, 72%) as a white solid. 1H NMR (300 MHz, DMSO-d6) δ 8.03 (d, J=1.1 Hz, 1H), 7.55 (dd, J=8.1, 2.1 Hz, 1H), 7.21 (d, J=8.1 Hz, 1H), 4.89 (s, 2H), 4.38 (s, 2H).

Step 5.

To a solution of compound 1.4 (506 mg, 2.77 mmol) in EtOH (9 mL) was added NaBH4 (104 mg, 2.77 mmol). The reaction was stirred at ambient temperature for 1 h. EtOAc (20 mL) and saturated aqueous NH4Cl (15 mL) was added to the reaction vessel and the resulting biphasic mixture was transferred to a separatory funnel. The layers were separated and the aqueous phase was extracted with EtOAc (3×20 mL). The combined organics were washed with brine, dried over anhydrous Na2SO4, filtered, and concentrated under reduced pressure to provide crude 6-chloroisochroman-4-ol 1.5 (478 mg, 94%) as colorless oil, which was used without further purification. 1H NMR (300 MHz, CDCl3) δ 7.47 (d, J=2.1 Hz, 1H), 7.25 (d, J=7.2 Hz, 1H), 6.98 (d, J=8.4 Hz, 1H), 4.79 (d, J=15.3 Hz, 1H), 4.66 (d, J=15.3 Hz, 1H), 4.55 (s, 1H), 4.08 (dd, J=12.0, 3.3 Hz, 1H), 3.88 (dd, J=12.0, 2.7 Hz, 1H), 3.73 (dd, J=14.1, 6.9 Hz, 1H), 2.36 (br s, 1H).

Step 6.

A solution of compound 1.5 (480 mg, 2.59 mmol) in toluene (7 mL) was treated with diphenyl phosphoryl azide (853 mg, 3.10 mmol), and cooled in an ice bath. A solution of DBU (777 mg, 3.10 mmol) in toluene (1.5 mL) was added, and the reaction was stirred at ambient temperature for 16 h. The reaction mixture was directly concentrated under reduced pressure and purified by flash column chromatography (SiO2, gradient elution from 5% EtOAc/hexanes to 10% EtOAc/hexanes) to afford 4-azido-6-chloroisochroman 1.6 (329 mg, 61%) as a colorless oil. 1H NMR (300 MHz, CDCl3) δ 7.39 (d, J=2.1 Hz, 1H), 7.32 (dd, J=8.1, 2.1 Hz, 1H), 7.04 (d, J=8.1 Hz, 1H), 4.86 (d, J=15.3 Hz, 1H), 4.70 (d, J=15.3 Hz, 1H), 4.24-2.19 (m, 2H), 3.97 (dd, J=11.4, 2.4 Hz, 1H).

Step 7.

A solution of compound 1.6 (330 mg, 1.57 mmol) in THF (15 mL) was treated with LiAlH4 (59.5 mg, 1.57 mmol) at room temperature. The reaction mixture was stirred at room temperature for 30 min. A solution of Na2CO3 (164 mg, 1.55 mmol) in water (10 mL), EtOAc (15 mL) and di-tert-butyl dicarbonate (0.676 g, 3.1 mmol) was added and the resulting mixture was stirred at ambient temperature for 4 h. The mixture was diluted with water (10 mL) was added to the reaction vessel and aqueous phase was extracted with EtOAc (3×25 mL). The combined organics were washed with brine, dried over anhydrous Na2SO4, filtered, and concentrated under reduced pressure. Purification by flash column chromatography (SiO2, gradient elution from 100% hexanes to 5% EtOAc/hexanes) gave tert-butyl (6-chloroisochroman-4-yl)carbamate 1.7 (349 mg, 78%) as a white solid. LC-MS (ESI): m/z 284 [M+H].

Step 8.

A solution of compound 1.7 (349 mg, 1.23 mmol) in Et2O (2 mL) was treated with a saturated solution of HCl in Et2O (5 mL). The reaction mixture was stirred at room temperature for 16 h and then concentrated under reduced pressure. The resulting solid residue was triturated with Et2O (2×5 mL) to provide the compound of Example 1 (165 mg, 61%) as a white solid. LC-MS (ESI): m/z 184 [M+H]. 1H NMR (300 MHz, DMSO-d6) δ 8.52 (brs, 3H), 7.64 (d, J=1.8 Hz, 1H), 7.46 (dd, J=8.4, 2.1 Hz, 1H), 7.24 (d, J=8.4 Hz, 1H), 4.83 (d, J=15.9 Hz, 1H), 4.68 (d, J=15.9 Hz, 1H), 4.38 (s, 1H), 4.14 (dd, J=12.3, 1.8 Hz, 1H), 3.89 (dd, J=12.6, 3.0 Hz, 1H).

TABLE 1 Compounds prepared as described in Example 1 using the appropriately substituted starting materials. Example Structure Characterization Data 2 MS (ESI): m/z 184 [M + H]. 1H NMR (300 M Hz, DMSO-d6) δ 8.58 (br s, 3H), 7.58 (d, J = 8.4 Hz, 1H), 7.42 (dd, J = 8.4, 2.1 Hz, 1H), 7.32 (d, J = 1.8 Hz, 1H), 4.83 (d, J = 15.6 Hz, 1H), 4.69 (d, J = 15.9 Hz, 1H), 4.36 (s, 1H), 4.18 (dd, J = 12.6, 1.8 Hz, 1H), 3.88 (dd, J = 12.6, 2.4 Hz, 1H). 3 MS (ESI): m/z 168 [M + H]. 1H NMR (300 M Hz, DMSO-d6) δ 8.61 (br s, 3H), 7.65-7.61 (m, 1H), 7.23-7.12 (m, 1H), 7.01 (dd, J = 9.6, 2.7 Hz, 1H), 4.83 (d, J = 15.9 Hz, 1H), 4.69 (d, J = 15.6 Hz, 1H), 4.35 (s, 1H), 4.20 (dd, J = 12.6, 1.8 Hz, 1H), 3.88 (dd, J = 12.6, 2.7 Hz, 1H). 4 MS (ESI): m/z 164 [M + H]. 1H NMR (300 M Hz, DMSO-d6) δ 8.50 (br s, 1H), 7.42 (d, J = 7.8 Hz, 1H), 7.14 (d, J = 7.8 Hz, 1H), 6.98 (s, 1H), 4.78 (d, J = 15.3 Hz, 1H), 4.67 (d, J = 15.3 Hz, 1H), 4.28 (s, 1H), 4.17 (dd, J = 12.3, 1.8 Hz, 1H), 3.87 (dd, J = 12.3, 2.7 Hz, 1H), 2.30 (s, 3H). 5 MS (ESI): m/z 184 [M + H]. 1H NMR (300 M Hz, DMSO-d6) δ 8.76 (s, 3H), 7.60 (d, J = 7.5 Hz, 1H), 7.51 (d, J = 7.2 Hz, 1H), 7.40 (t, J = 7.8 Hz, 1H), 4.82 (d, J = 15.9 Hz, 1H), 4.63 (d, J = 15.9 Hz, 1H), 4.39 (s, 1H), 4.24 (d, J = 12.3 Hz, 1H), 3.89 (dd, J = 12.6, 2.4 Hz, 1H). 6 MS (ESI): m/z 167 [M + H]. 1H NMR (300 M Hz, DMSO-d6) δ 8.57 (br s, 2H), 7.43 (d, J = 9.6 Hz, 1H), 7.24 (d, J = 12.0 Hz, 2H), 4.82 (d, J = 15.3 Hz, 1H), 4.68 (d, J = 15.3 Hz, 1H), 4.37 (s, 1H), 4.14 (d, J = 12.6 Hz, 1H) 3.90 (dd, J = 12.6 Hz, 3.0 Hz, 1H).

Example 7: 6-Phenylisochroman-4-amine hydrochloride

Step 1.

A solution of 170 mg (0.45 mmol) of tert-butyl (6-iodoisochroman-4-yl)carbamate 7.1 (prepared from methyl 2-(bromomethyl)-5-iodobenzoate (Hayakawa, I., et al, WO2011068171) as described in Example 1) in 1,4-dioxane (4 mL) was treated with phenyl boronic acid (110 mg, 0.90 mmol) and Pd(PPh3)4(52.3 mg, 0.045 mmol) and stirred at room temperature for 15 min. Aqueous 3M K2CO3 (0.76 mL, 2.3 mmol) was added, and the reaction mixture was heated to 100° C. with stirring for 5 h. After cooling to room temperature, the mixture was diluted with water (5 mL) and extracted with EtOAc (3×15 mL). The combined organics were dried over anhydrous Na2SO4, filtered, and concentrated under reduced pressure. Purification by flash column chromatography (SiO2, gradient elution from 1% EtOAc/hexanes to 5% EtOAc/hexanes) gave tert-butyl (6-phenylisochroman-4-yl) carbamate 7.2 (110 mg, 75%) as a white solid. LC-MS (ESI): m/z 326 [M+H].

Step 2.

The compound of Example 7 was prepared as previously described in Example 1. MS (ESI): m/z 226 [M+H]. 1HNMR (300 MHz, DMSO) δ 8.50 (s, 3H), 7.87 (s, 1H), 7.11 (d, J=7.5 Hz, 3H), 7.53-7.37 (m, 3H), 7.28 (d, J=8.1 Hz, 1H), 4.88 (d, J=15.9 Hz, 1H), 4.75 (d, J=15.3 Hz, 1H), 4.42 (s, 1H), 4.17 (d, J=12.6 Hz, 1H), 3.95 (dd, J=12.3, 2.4 Hz, 1H).

Example 8: 6-Isopropylisochroman-4-amine hydrochloride

Step 1.

10% Pd/C (25 mg) was added to 250 mg (0.86 mmol) of tert-butyl (6-(prop-1-en-2-yl)isochroman-4-yl)carbamate 8.1 (prepared as described in Example 7 using isopropenylboronic acid pinacol ester) in CH3OH (25 mL). The reaction mixture was stirred at room temperature under a H2 atmosphere for 16 h. After filtering through a Celite pad, the filtrate was concentrated under reduced pressure. Purification by flash column chromatography (SiO2, 5% EtOAc/hexanes) afforded tert-butyl (6-isopropylisochroman-4-yl) carbamate 8.2 (180 mg, 72%) as a brown oil. 1H NMR (300 MHz, CDCl3) δ 7.29 (s, 1H), 7.15 (dd, J=8.1, 1.5 Hz, 1H), 6.95 (d, J=8.1 Hz, 1H), 5.10 (d, J=9.0 Hz, 1H), 4.81-4.68 (m, 3H), 4.07 (dd, J=9.0, 1.8 Hz, 1H), 3.87 (dd, J=12.0, 3.0 Hz, 1H), 2.95-2.86 (m, 1H), 1.60 (s, 9H), 0.97 (d, J=7.2 Hz, 6H).

Step 2.

The compound of Example 8 was prepared as described in Example 1. MS (ESI): m/z 192 [M+H]. 1HNMR (300 MHz, DMSO-d6) δ 8.38 (br s, 3H), 7.42 (s, 1H), 7.27 (d, J=8.1 Hz, 1H), 7.09 (d, J=8.1 Hz, 1H), 4.79 (d, J=15.0 Hz, 1H), 4.67 (d, J=15.6 Hz, 1H), 4.31 (s, 1H), 4.14 (d, J=12.3 Hz, 1H), 3.87 (d, J=12.6 Hz, 1H), 2.91-2.86 (m, 1H), 1.21 (d, J=6.9 Hz, 6H).

Example 9: 6-Trifluoromethylisochroman-4-amine hydrochloride

Step 1.

A solution of compound 7.1 (227 mg, 605 μmol) in anhydrous DMF (8 mL) under a N2 atmosphere was treated with copper(I) iodide (230 mg, 1.21 mmol) and hexamethylphosphoramide (541 mg, 3.02 mmol), and stirred at ambient temperature for 10 min. Methyl 2,2-difluoro-2-(fluorosulfonyl)acetate (580 mg, 3.02 mmol) was added, and the reaction mixture was heated to 80° C. and stirred for 6 h. After cooling to room temperature, the mixture was diluted with saturated aqueous NaHCO3 (20 mL) and partitioned with EtOAc (20 mL). The aqueous phase was extracted with EtOAc (2×30 mL). The combined organics were dried over anhydrous Na2SO4, filtered, and concentrated under reduced pressure. Purification by flash column chromatography (SiO2, gradient elution from 5% EtOAc/hexanes to 10% EtOAc/hexanes) afforded tert-butyl (6-(trifluoromethyl) isochroman-4-yl)carbamate 9.1 (85.0 mg, 44%) as a white solid. MS (ESI): m/z 340 [M+Na].

Step 2.

The compound of Example 9 was prepared as previously described in Example 1. MS (ESI): m/z 218 [M+H]. 1HNMR (300 MHz, DMSO-d6) δ 8.57 (s, 3H), 7.96 (s, 1H), 7.75 (d, J=7.5 Hz, 1H), 7.44 (d, J=8.1 Hz, 1H), 4.92 (d, J=16.2 Hz, 1H), 4.77 (d, J=15.6 Hz, 1H), 4.95 (s, 1H), 4.18 (d, J=12.3 Hz, 1H), 3.93 (dd, J=12.6, 2.4 Hz, 1H).

Example 10: 6-Methoxyisochroman-4-amine hydrochloride

Step 1.

To a solution of prop-2-en-1-ol (6.38 g, 110 mmol) in DMF (400 mL) was added 95% sodium hydride (4.38 g, 110 mmol) and 2-bromo-1-(bromomethyl)-4-methoxybenzene (15.5 g, 55.3 mmol) at 0° C. The resulting suspension was stirred at room temperature for 30 min. Excess hydride was quenched by the dropwise addition of water (200 mL), and the mixture was extracted with EtOAc (3×500 mL). The combined organics were washed with brine (2×300 mL), dried over anhydrous Na2SO4, filtered, and concentrated under reduced pressure. The resulting oil was purified by flash column chromatography (SiO2, gradient elution from 100% hexanes to 5% EtOAc/hexanes) to provide 1-((allyloxy)methyl)-2-bromo-4-methoxy benzene 10.1 (12.9 g, 91%) as a yellow oil. 1HNMR (300 MHz, CDCl3) δ 7.42 (d, J=8.7 Hz, 1H), 7.10 (d, J=3.3 Hz, 1H), 6.72 (dd, J=8.7, 3.3 Hz, 1H), 6.05-5.94 (m, 1H), 5.40 (ddd, J=17.1, 3.3, 1.5 Hz, 1H), 5.25 (dd, J=10.5, 1.5 Hz, 1H), 4.57 (s, 2H), 4.14 (m, 2H), 3.82 (s, 3H).

Step 2.

A solution of compound 10.1 (12.9 g, 50.1 mmol) in DMF (400 mL) was added Cs2CO3 (19.5 g, 60.1 mmol), triphenylphosphine (5.90 g, 22.5 mmol) and Pd(OAc)2 (1.68 g, 7.51 mmol). The reaction mixture was heated to 90° C. for 1 h. Water (300 mL) and EtOAc (300 mL) were added to the reaction vessel and the resulting biphasic mixture was transferred to a separatory funnel. The layers were separated and the aqueous phase was extracted with EtOAc (2×500 mL). The organic phase was washed with brine (3×500 mL). The combined organics were dried over anhydrous Na2SO4, filtered, and concentrated under reduced pressure. The resulting oil was purified by flash column chromatography (SiO2, gradient elution from 100% hexanes to 5% EtOAc/hexanes) to afford 6-methoxy-4-methylene isochroman 10.2 (5.70 g, 64%) as a yellow oil. 1H NMR (300 MHz, CDCl3) δ 7.64 (d, J=8.7 Hz, 1H), 6.82 (dd, J=7.2, 2.7 Hz, 1H), 6.56 (d, J=2.7 Hz, 1H), 5.48 (s, 1H), 4.91 (s, 1H), 4.80 (s, 2H), 4.44 (s, 2H), 3.82 (s, 3H).

Step 3.

NaIO4 (20.7 g, 96.8 mmol) and OsO4 (81.8 mg, 322 μmol) were added to a solution of 12.2 (5.7 g, 32.3 mmol) in THF (200 mL) and water (100 mL), and the reaction mixture was stirred at room temperature for 4 h. The mixture was diluted with water (200 mL) and extracted with EtOAc (3×200 mL). The combined organics were dried over anhydrous Na2SO4, filtered, and concentrated under reduced pressure. The resulting oil was purified by flash column chromatography (SiO2, gradient elution from 5% EtOAc/hexanes to 10% EtOAc/hexanes) to provide 6-methoxyisochroman-4-one 10.3 (3.10 g, 54%) as a white solid. 1HNMR (300 MHz, CDCl3) δ 7.53 (s, 1H), 7.27 (d, J=6.6 Hz, 1H), 7.16 (d, J=1.5 Hz, 1H), 4.87 (s, 2H), 4.37 (s, 2H), 3.88 (s, 3H).

Step 4.

6-Methoxyisochroman-4-ol 10.4 (3.00 g, 96%) was prepared as described in Example 1. 1H NMR (300 MHz, CDCl3) δ 7.03-6.98 (m, 2H), 6.88 (m, 1H), 4.79 (A of AB, JAB=14.7 1H), 4.66 (B of AB, JBA=14.7 1H), 4.54 (m, 1H), 4.12 (A of ABX, JAB=12.0 Hz, JAX=2.7 Hz, 1H), 3.88 (B of ABX, JBA=12.0 Hz, JBX=2.7 Hz, 1H), 3.80 (s, 3H), 2.23 (d, J=9.6 Hz, 1H).

Step 5.

4-Azido-6-methoxyisochroman 10.5 (2.70 g, 79%) was prepared as described in Example 1. 1H NMR (300 MHz, CDCl3) δ 7.01 (d, J=8.1 Hz, 1H), 6.94-6.90 (m, 2H), 4.84 (A of AB, JAB=14.7 Hz, 1H), 4.69 (B of AB, JBA=14.7 Hz, 1H), 4.33-3.97 (m, 2H), 3.92 (dd, J=12.9, 2.4 Hz, 1H), 3.85 (s, 3H).

Step 6.

tert-Butyl (6-methoxyisochroman-4-yl)carbamate 10.6 (2.49 g, 68%) was prepared as described in Example 1. LC-MS (ESI): m/z 316 [M+H].

Step 7.

The compound of Example 10 (90.0 mg, 94%) was prepared as described in Example 1. MS (ESI): m/z 180 [M+H]. 1H NMR (300 MHz, DMSO-d6) δ 8.50 (s, 3H), 7.16 (d, J=2.1 Hz, 1H), 7.10 (d, J=8.4 Hz, 1H), 6.97 (dd, J=8.4, 2.4 Hz, 1H), 4.76 (d, J=14.7 Hz, 1H), 4.64 (d, J=15.0 Hz, 1H), 4.31 (s, 1H), 4.12 (d, J=12.9 Hz, 1H), 3.88 (dd, J=12.3, 2.7 Hz, 1H), 3.77 (s, 3H).

TABLE 2 Compounds prepared as described in Example 10. Example Structure Characterization Data 11 MS (ESI): m/z 206 [M + H]. 1H NMR (300 M Hz, DMSO-d6) δ 8.53 (br s, 3H), 7.60 (s, 1H), 7.41 (dd, J = 8.1, 1.8 Hz, 1H), 7.09 (d, J = 8.1 Hz, 1H), 4.79 (d, J = 15.0 Hz, 1H), 4.66 (d, J = 15.3 Hz, 1H), 4.31 (s, 1H), 4.15 (d, J = 12.3 Hz, 1H), 3.89 (dd, J = 12.6, 2.7 Hz, 1H), 1.29 (s, 9H). 12 MS (ESI): m/z 150 [M + H]. 1H NMR (300 M Hz, CD3OD) δ 7.48-7.34 (m, 3H), 7.20 (d, J = 7.5 Hz, 1H), 4.95 (d, J = 15.3 Hz, 1H), 4.81 (d, J = 15.6 Hz, 1H), 4.38 (s, 1H), 4.26 (dd, J = 12.9, 1.5 Hz, 1H), 3.99 (dd, J = 10.5, 2.1 Hz, 1H). 13 MS (ESI): m/z 200 [M + H]. 1H NMR (300 M Hz, DMSO-d6) δ 8.46 (br s, 3H), 8.17 (d, J = 8.4 Hz, 1H), 7.99 (d, J = 8.4 Hz, 2H), 7.65-7.60 (m, 2H), 7.31 (d, J = 8.4 Hz, 1H), 5.05-4.88 (m, 3H), 4.38 (d, J = 12.3 Hz, 1H), 3.96 (dd, J = 12.6, 1.8 Hz, 1H). 14 MS (ESI): m/z 180 [M + H]. 1H NMR (300 M Hz, CD3OD) δ 7.36 (d, J = 8.4 Hz, 1H), 6.91 (dd, J = 8.4, 2.7 Hz, 1H), 6.75 (d, J = 2.1 Hz, 1H), 4.90 (d, J = 15.6 Hz, 1H), 4.75 (d, J = 15.6 Hz, 1H), 4.30 (s, 1 H), 4.22 (dd, J = 12.6, 1.2 Hz, 1H), 3.94 (dd, J = 12.6, 2.1 Hz, 1H), 3.82 (s, 3H). 15 MS (ESI): m/z 201 [M + H]. 1H NMR (300 M Hz, CD3OD) δ 9.43 (d, J = 8.7 Hz, 1H), 9.29 (d, J = 5.1 Hz, 1H), 8.32 (d, J = 9.0 Hz, 1H), 8.22 (dd, J = 8.7, 5.1 Hz, 1H), 7.99 (d, J = 9.0 Hz, 1H), 5.25 (d, J = 17.7 Hz, 2H), 5.11 (t, J = 17.7 Hz, 1H), 4.50 (d, J = 12.9 Hz, 1H), 4.12 (d, J = 14.7 Hz, 1H). 16 MS (ESI): m/z 221 [M + H]. 1H NMR (300 M Hz, DMSO-d6) δ 8.04 (d, J = 8.4 Hz, 1H), 7.22 (d, J = 8.4 Hz, 1H), 4.95 (d, J = 15.6 Hz, 1H), 4.79 (d, J = 16.2 Hz, 2H), 4.23 (d, J = 12.3 Hz, 1H), 4.00 (dd, J = 12.6, 2.4 Hz, 1H), 2.85 (s, 3H).

Example 17: 8-Ethylisochroman-4-amine hydrochloride

Step 1.

tert-Butyl (8-vinylisochroman-4-yl)carbamate 17.2 was prepared from compound 17.1 (prepared from 1,3-dibromo-2-(bromomethyl)benzene as previously described in Example 10) as previously described in Example 7 using vinylboronic acid pinacol ester. 1H NMR (300 MHz, CDCl3) δ 7.40-7.35 (m, 2H), 7.27-7.22 (m, 1H), 6.60 (dd, J=18.0, 12.0 Hz, 1H), 5.68 (d, J=18.0 Hz, 1H), 5.36-5.30 (m, 1H), 5.11-5.08 (m, 1H), 4.90 (d, J=15.0 Hz, 1H), 4.75-4.67 (m, 2H), 4.03 (dd, J=12.0, 3.0 Hz, 1H), 3.83 (dd, J=12.0, 3.0 Hz, 1H), 1.46 (s, 9H).

Step 2.

tert-Butyl (8-ethylisochroman-4-yl)carbamate 17.3 was prepared as previously described in Example 8. 1H NMR (300 MHz, CDCl3) δ 7.29-7.19 (m, 2H), 7.13-7.10 (m, 1H), 5.10-5.07 (m, 1H), 4.86 (d, J=15.0 Hz, 1H), 4.77-4.71 (m, 2H), 4.04 (dd, J=12.0, 3.0 Hz, 1H), 3.86 (dd, J=12.0, 3.0 Hz, 1H), 2.51-2.44 (m, 2H), 1.46 (s, 9H), 1.20 (t, J 9.0 Hz, 3H).

Step 3.

The compound of Example 17 was prepared as previously described in Example 1. MS (ESI): m/z 178 [M+H]. 1H NMR (300 MHz, CD3OD) δ 7.31-7.27 (m, 3H), 5.00 (d, J=18.0 Hz, 1H), 4.76 (d, J=15.0 Hz, 1H), 4.33 (s, 1H), 4.20 (dd, J=15.0, 3 Hz, 1H), 3.92 (dd, J=12.0, 3 Hz, 1H), 2.58-2.50 (m, 2H), 1.21 (t, J=9.0 Hz, 3H).

Example 18: 8-Isopropylisochroman-4-amine hydrochloride

The compound of Example 18 was prepared as described in Example 17 using isopropenylboronic acid pinacol ester. MS (ESI): m/z 192 [M+H]. 1H NMR (300 MHz, CD3OD) δ 7.41-7.31 (m, 2H), 7.26-7.23 (m, 1H), 5.08 (d, J=18 Hz, 1H), 4.81 (d, J=21 Hz, 1H), 4.32 (s, 1H), 4.19 (dd, J=12.6, 1.2 Hz, 1H), 3.92 (dd, J=12.9, 2.1 Hz, 1H), 2.86-2.48 (m, 1H), 1.26-1.20 (m, 6H).

Example 19: 2,4,7,8,9,10-Hexahydro-1H-benzo[f]isochromen-1-amine hydrochloride

The compound of Example 19 was prepared as described in Example 10 using 5-bromo-6-(bromomethyl)-1,2,3,4-tetrahydronaphthalene 19.6. MS (ESI): m/z 204 [M+H]. 1H NMR (300 MHz, DMSO-d6) δ 8.38 (s, 3H), 7.09 (d, J=7.8 Hz, 1H), 6.89 (d, J=7.8 Hz, 1H), 4.82 (d, J=15.6 Hz, 1H), 4.67 (d, J=15.6 Hz, 1H), 4.36 (d, J=3.9 Hz, 1H), 4.23 (d, J=12.3 Hz, 1H), 3.75 (d, J=12.0 Hz, 1H), 2.92-2.84 (m, 1H), 2.73-2.65 (m, 3H), 1.83-1.60 (m, 4H).

Synthesis of 5-bromo-6-(bromomethyl)-1,2,3,4-tetrahydronaphthalene (19.6)

Step 1.

Nitric acid (10.7 mL, 240 mmol) was added to a solution of 5,6,7,8-tetrahydronaphthalene-2-carboxylic acid (10.0 g, 56.7 mmol) in AcOH (70 mL and concentrated H2SO4 (70 mL). The reaction was stirred at ambient temperature for 1 h and then poured onto ice. The resulting precipitate was collected by filtration to provide 1-nitro-5,6,7,8-tetrahydronaphthalene-2-carboxylic acid 19.1 (11.2 g, 89%) as a yellow solid. MS (ESI−): m/z 220 [M−H]. 1H NMR (300 MHz, DMSO-d6) δ 7.75 (d, J=8.1 Hz, 1H), 7.41 (d, J=8.1 Hz, 1H), 2.88-2.81 (m, 2H), 2.69-2.63 (m, 2H), 1.74 (m, 4H).

Step 2.

A solution of compound 19.1 (5.2 g, 11.7 mmol) in CH3CN (100 mL) was treated with K2CO3 (4.85 g, 35.1 mmol) and iodomethane (4.98 g, 35.1 mmol). The reaction was stirred at room temperature for 3 h. The mixture was then partitioned between water (100 mL) and EtOAc (600 mL), and the organic phase was washed with brine (2×100 mL), dried over anhydrous Na2SO4, filtered, and concentrated under reduced pressure. The resulting oil was purified by flash column chromatography (SiO2, 100% hexanes) to provide methyl 1-nitro-5,6,7,8-tetrahydronaphthalene-2-carboxylate 19.2 (2.24 g, 81%) as a white solid. 1H NMR (300 MHz, CDCl3) δ 7.77 (d, J=8.1 Hz, 1H), 7.26 (d, J=8.4 Hz, 1H), 3.89 (s, 3H), 2.87-2.85 (m, 2H), 2.72-2.66 (m, 2H), 1.83-1.81 (m, 4H).

Step 3.

Compound 19.2 (3.2 g, 9.52 mmol) was dissolved in THF (100 mL). Pd/C (1 g, 10%) was added and the mixture was stirred at room temperature under a H2 atmosphere for 8 h. The mixture was filtered through a Celite pad, and the filtrate was concentrated under reduced pressure. Purification by flash column chromatography (SiO2, 100% hexanes) gave methyl 1-amino-5,6,7,8-tetrahydronaphthalene-2-carboxylate 19.3 (1.80 g, 92%) as a white solid. 1H NMR (300 MHz, CDCl3) δ 7.65 (d, J=8.4 Hz, 1H), 6.42 (d, J=8.4 Hz, 1H), 5.88 (brs, 2H), 3.85 (s, 3H), 2.72 (t, J=6.3 Hz, 2H), 2.42 (t, J=6.0 Hz, 2H), 1.92-1.84 (m, 2H), 1.78-1.71 (m, 2H).

Step 4.

A solution of 19.3 (4.3 g, 20.9 mmol) in CH3CN (200 mL) was cooled in an ice bath, treated with copper(I) bromide (5.99 g, 41.8 mmol), and stirred for 30 min. tert-Butyl nitrite (4.31 g, 41.8 mmol) was added, and the ice bath was removed. The flask was heated to 40° C. and stirred overnight. The mixture was cooled in an ice bath and diluted with NH4OH and EtOAc, and the resulting biphasic mixture was separated. The aqueous phase was extracted with EtOAc (3×100 mL). The combined organics were dried over anhydrous Na2SO4, filtered, and concentrated under reduced pressure. Purification by flash column chromatography (SiO2, 5% EtOAc/hexanes) gave methyl 1-bromo-5,6,7,8-tetrahydronaphthalene-2-carboxylate 19.4 (3.60 g, 63%) as a yellow solid. 1H NMR (300 MHz, CDCl3) δ 7.40 (d, J=7.8 Hz, 1H), 7.06 (d, J=7.8 Hz, 1H), 3.91 (s, 3H), 2.80 (t, J=8.1 Hz, 4H), 1.86-1.72 (m, 4H).

Step 5.

Compound 19.4 (3.6 g, 13.3 mmol) was dissolved in anhydrous THF (100 mL) under a N2 atmosphere and cooled to −78° C. in a dry ice/acetone bath. Diisobutylaluminum hydride (17.7 mL, 26.6 mmol, 1.5M in toluene) was added in a dropwise manner. Once addition was complete, the cold bath was removed and the reaction mixture was allowed to warm to room temperature for 1 h. Excess hydride was carefully quenched by the dropwise addition of aqueous 3M HCl (50 mL) and the biphasic mixture was transferred to a separatory funnel. The aqueous phase was washed with EtOAc (3×150 mL). The combined organics were dried over anhydrous Na2SO4, filtered, and concentrated under reduced pressure. Purification by flash column chromatography (SiO2, 10% EtOAc/hexanes) gave (1-bromo-5,6,7,8-tetrahydronaphthalen-2-yl)methanol 19.5 (3.0 g, 94%) as a white solid. 1H NMR (300 MHz, CDCl3) δ 7.20 (d, J=7.8 Hz, 1H), 7.06 (d, J=7.8 Hz, 1H), 4.73 (d, J=6.6 Hz, 2H), 2.80-2.75 (m, 4H), 2.02 (t, J=6.6 Hz, 1H), 1.88-1.71 (m, 4H).

Step 6.

Compound 19.5 (3.0 g, 12.4 mmol) was dissolved in 1,2-dichloroethane (120 mL) and cooled in an ice bath. Phosphorous tribromide (0.59 mL, 6.20 mmol) was added and the mixture was stirred at 0° C. for 30 min. The reaction poured into ice water and extracted with CH2Cl2 (3×50 mL). The combined organics were washed with saturated aqueous Na2CO3 (100 mL), dried over anhydrous Na2SO4, filtered, and concentrated under reduced pressure to afford 5-bromo-6-(bromomethyl)-1,2,3,4-tetrahydronaphthalene 19.6 (1.70 g, 45%). 1H NMR (300 MHz, CDCl3) δ 7.10 (d, J=7.8 Hz, 1H), 7.02 (d, J=7.5 Hz, 1H), 4.65 (s, 2H), 2.80-2.71 (m, 4H), 1.87-1.70 (m, 4H).

Example 20: 6-Ethylisochroman-4-amine hydrochloride

The compound of Example 20 was prepared as described in Example 19 using methyl 2-amino-4-ethylbenzoate. MS (ESI): m/z 178 [M+H]. 1H NMR (300 MHz, DMSO-d6) δ 8.44 (s, 3H), 7.35 (s, 1H), 7.23 (d, J=7.8 Hz, 1H), 7.09 (d, J=8.1 Hz, 1H), 4.84 (d, J=11.7 Hz, 1H), 4.70 (d, J=15.3 Hz, 1H), 4.32 (s, 1H), 4.16 (dd, J=12.6, 1.8 Hz, 1H), 3.90 (dd, J=12.6, 2.7 Hz, 1H), 2.64-2.57 (m, 2H), 1.24-1.09 (m, 3H).

Example 21: 5-Methylisochroman-4-amine hydrochloride

Step 1.

Sodium hydride (771 mg, 19.3 mmol, 95%) was added in small portions to a solution of (2-bromo-3-methylphenyl)methanol (2.6 g, 12.9 mmol) in DMF (35 mL) cooled in an ice bath. The resulting slurry was stirred for 15 min, followed by addition of 3-bromoprop-1-ene (1.70 g, 14.1 mmol). The reaction was stirred at 0° C. for 30 min. Excess hydride was quenched by the dropwise addition of saturated aqueous NH4Cl (75 mL), and the mixture was partitioned with Et2O (3×40 mL). The combined organics were dried over anhydrous Na2SO4, filtered, and concentrated under reduced pressure. The resulting oil was purified by flash column chromatography (SiO2, 100% hexanes) to provide 1-((allyloxy)methyl)-2-bromo-3-methylbenzene 21.1 (2.30 g, 49%) as a colorless oil. 1H NMR (300 MHz, CDCl3) δ 7.36-7.34 (m, 1H), 7.28-7.18 (m, 2H), 6.08-5.95 (m, 1H), 5.38 (d, J=15.0 Hz, 1H), 5.25 (d, J=10.5 Hz, 1H), 4.62 (s, 2H), 4.14 (d, J=5.7 Hz, 2H), 2.44 (s, 3H).

Step 2.

5-Methyl-4-methyleneisochroman 21.2 was prepared as previously described in Example 10. 1H NMR (300 MHz, CDCl3) δ 7.19-7.15 (m, 2H), 6.96-6.92 (m, 1H), 5.47 (s, 1H), 5.37 (s, 1H), 4.81 (s, 2H), 4.41 (s, 2H), 2.53 (s, 3H).

Step 3.

5-Methylisochroman-4-one 21.3 was prepared as previously described in Example 10. 1H NMR (300 MHz, CDCl3) δ 7.44 (t, J=7.5 Hz, 1H), 7.21 (d, J=7.5 Hz, 1H), 7.08 (d, J=7.8 Hz, 1H), 4.88 (s, 2H), 4.35 (s, 2H), 2.70 (s, 3H).

Step 4.

5-Methylisochroman-4-ol 21.4 was prepared as previously described in Example 1. 1H NMR (300 MHz, CDCl3) δ 7.24-7.19 (m, 1H), 7.13 (d, J=7.5 Hz, 1H), 6.88 (d, J=7.5 Hz, 1H), 4.83 (d, J=15.0 Hz, 1H), 4.70 (d, J=15.0 Hz, 1H), 4.60 (d, J=10.2 Hz, 1H), 4.29 (d, J=12.0 Hz, 1H), 3.81 (dd, J=12.0, 3.3 Hz, 1H), 2.48 (s, 3H), 2.23 (dd, J=13.5, 7.5 Hz, 1H).

Step 5.

4-Azido-5-methylisochroman 21.5 was prepared as previously described in Example 1. 1H NMR (300 MHz, CDCl3) δ 7.26-7.24 (m, 1H), 7.18 (d, J=7.2 Hz, 1H), 6.94 (d, J=7.2 Hz, 1H), 4.91 (d, J=15.3 Hz, 1H), 4.74 (d, J=15.3 Hz, 1H), 4.28 (d, J=12.0 Hz, 1H), 4.08 (s, 1H), 3.92 (dd, J=12.3, 2.1 Hz, 1H), 2.42 (s, 3H).

Step 6.

The compound of Example 21 was prepared as previously described in Example 1. MS (ESI): m/z 164 [M+H]. 1H NMR (300 MHz, DMSO-d6) δ 8.26 (s, 3H), 7.29 (t, J=7.5 Hz, 1H), 7.15 (d, J=7.2 Hz, 1H), 6.99 (d, J=7.8 Hz, 1H), 4.87 (d, J=15.6 Hz, 1H), 4.72 (d, J=15.6 Hz, 1H), 4.43 (s, 1H), 4.21 (d, J=12.3 Hz, 1H), 3.80 (dd, J=12.3, 1.2 Hz, 1H), 2.39 (s, 3H).

TABLE 3 Compounds prepared as described in Example 21 using the appropriately substituted starting material. Example Structure Characterization Data 22 MS (ESI): m/z 168 [M + H]. 1H NMR (300 M Hz, CD3OD) δ 7.52-7.44 (m, 1H), 7.14 (t, J = 9.0 Hz, 1H), 7.06 (d, J = 7.8 Hz, 1H), 4.97 (d, J = 15.9 Hz, 1H), 4.80 (d, J = 15.9 Hz, 1H), 4.58 (s, 1H), 4.28 (d, J = 12.9 Hz, 1H), 3.95 (dd, J = 13.2, 2.4 Hz, 1H). 23 MS (ESI): m/z 168 [M + H]. 1H NMR (300 M Hz, CD3OD) δ 7.46-7.39 (m, 1H), 7.30 (d, J = 7.8 Hz, 1H), 7.20 (t, J = 9.3 Hz, 1H), 5.05 (d, J = 15.9 Hz, 1H), 4.77 (d, J = 15.9 Hz, 1H), 4.41 (s, 1H), 4.24 (d, J = 12.0 Hz, 1H), 3.97 (dd, J = 12.9, 2.1 Hz, 1H). 24 MS (ESI): m/z 180 [M + H]. 1H NMR (300 M Hz, CD3OD) δ 7.37 (t, J = 6.0 Hz, 1H), 7.05 (d, J = 6.3 Hz, 1H), 4.97 (d, J = 12.3 Hz, 1H), 4.65 (d, J = 12.0 Hz, 1H), 4.34 (s, 1 H), 4.21 (dd, J = 9.6, 0.9 Hz, 1H), 3.93 (dd, J = 9.6, 1.5 Hz, 1H), 3.88 (s, 3H). 25 MS (ESI): m/z 180 [M + H]. 1H NMR (300 M Hz, CD3OD) δ 7.38 (t, J = 7.8 Hz, 1H), 6.96 (d, J = 7.5 Hz, 1H), 6.77 (d, J = 7.8 Hz, 1H), 4.89 (d, J = 15.0 Hz, 1H), 4.72 (d, J = 15.3 Hz, 1H), 4.44 (d, J = 1.2 Hz, 1 H), 4.25 (d, J = 12.9 Hz, 1H), 3.93 (s, 3H), 3.90 (dd, J = 13.5, 3.0 Hz, 1H). 26 MS (ESI): m/z 164 [M + H]. 1H NMR (300 M Hz, DMSO-d6) δ 8.69 (brs, 3H),7.32 (s, 1H) , 7.21 (d, J = 7.8 Hz, 1H), 7.07 (d, J = 7.8 Hz, 1H), 4.80 (d, J = 15.3 Hz, 1H), 4.67 (d, J = 15.3 Hz, 1H), 4.29 (s, 1H), 4.13 (dd, J = 12.6, 1.8 Hz, 1H), 3.88 (dd, J = 12.6, 2.7 Hz, 1H), 2.30 (s, 3H). 27 MS (ESI): m/z 164 [M + H]. 1H NMR (300 M Hz, CD3OD) δ 7.29-7.25 (m, 3H), 4.94 (d, J = 17.7 Hz, 1H), 4.71 (d, J = 15.9 Hz, 1H), 4.35 (s, 1H), 4.23 (d, J = 12.6 Hz, 1H), 3.95(dd, J = 12.6, 1.8 Hz, 1H), 2.21 (s, 3H). 28 MS (ESI): m/z 184 [M + H]. 1H NMR (300 M Hz, DMSO-d6) δ 8.49 (s, 3H), 7.48-7.42 (m, 2H), 7.20 (d, J = 6.3 Hz, 1H), 4.89 (d, J = 15.9 Hz, 1H), 4.71 (d, J = 15.6 Hz, 1H), 4.48 (s, 1H), 4.28 (d, J = 12.6 Hz, 1H), 3.76 (d, J = 12.6 Hz, 1H). 29 MS (ESI): M/Z 178 [M + H]. 1H NMR (300 M Hz, CDCl3) δ 8.78 (brs, 3 H), 7.61 (d, J = 7.5 Hz, 1H), 7.35 (t, J = 7.5 Hz, 1H), 7.26-7.21 (m, 1H), 7.15 (d, J = 7.8 Hz, 1H), 4.36 (s, 1H), 4.29 (d, J = 12 Hz, 1H), 4.04 (d, J = 12 Hz, 1H), 1.78 (s, 3H), 1.50 (s, 3H). 30 MS (ESI): m/z 164 [M + H]. 1H NMR (300 M Hz, DMSO-d6) δ 8.59 (brs, 3H), 7.36-7.23 (m, 4H), 4.41 (s, 1H), 4.12-4.00 (m, 2H), 3.74 (d, J = 12.6 Hz, 1H), 3.60-3.49 (m, 2H), 2.89- 2.80 (m, 1H). 32 MS (ESI): m/z 214 [M + H]. 1H NMR (300 M Hz, CD3OD) δ 8.23 (d, J = 8.7 Hz, 1H), 7.94 (t, J = 8.4 Hz, 2H), 7.66 (t, J = 7.2 Hz, 1H), 7.56 (t, J = 7.5 Hz, 1H), 7.41 (d, J = 8.4 Hz, 1H), 5.52 (d, J = 4.2 Hz, 1H), 4.46-4.31 (m, 2H), 3.90 (d, J = 14.4 Hz, 1H), 3.84-3.70 (m, 2H), 3.10-3.04 (m, 1H).

Example 31: 8-Methyl-1,2,4,5-tetrahydrobenzo[d]oxepin-1-amine hydrochloride

The compound of Example 31 was prepared as previously described in Example 21 using 2-(2-bromo-4-methylphenyl)ethanol. MS (ESI): m/z 178 [M+H]. 1H NMR (300 MHz, CDCl3) δ 7.71 (s, 1H), 7.08-7.01 (m, 2H), 4.48 (d, J=11.4 Hz, 1H), 4.37 (s, 1H), 4.16 (d, J=12.6 Hz, 1H), 3.77-3.72 (m, 2H), 3.54 (t, J=11.7 Hz, 1H), 2.77-2.70 (m, 1H), 1.61 (s, 3H).

Synthesis of 2-(2-bromo-4-methylphenyl)ethanol

Step 1.

A solution of 2-(2-bromo-4-methylphenyl)acetic acid (7.2 g, 31.43 mmol) in THF (314 mL) was cooled in an ice bath to 0° C., and treated with BH3.THF (62.86 mL, 62.86 mmol, 1M in THF) in a dropwise manner. After addition was complete, the reaction mixture was stirred at 0° C. overnight. Excess borane was quenched by the dropwise addition of 1M HCl (20 mL) at 0° C. The mixture was concentrated under reduced pressure and then partitioned between EtOAc (300 mL) and water (200 mL). The organic layer was washed with brine (200 mL), dried over anhydrous Na2SO4, filtered, and concentrated to give the 2-(2-bromo-4-methylphenyl)ethanol (6.7 g, 94%) as a yellow oil. MS (ESI) m/z 214, 216 [M+H].

Example 33: 1,3,4,5-Tetrahydrobenzo[c]oxepin-5-amine hydrochloride

The compound of Example 33 was prepared as previously described in Example 21 using 4-bromobut-1-ene in place of 3-bromoprop-1-ene. MS (ESI): m/z 164 [M+H]. 1H NMR (300 MHz, CD3OD) δ 7.43-7.29 (m, 4H), 4.90-4.76 (m, 3H), 4.22-4.14 (m, 1H), 4.09-4.03 (m, 1H), 2.28-2.20 (m, 2H)

TABLE 4 Compounds prepared as described in Example 33 using the appropriately substituted 2-bromobenzyl alcohol. Example Structure Characterization Data 34 MS (ESI+): m/z 178 [M + H]. 1H NMR (300 M Hz, CD3OD) δ 7.18-7.13 (m, 3H), 4.88-4.71 (m, 3H), 4.20-4.15 (m, 1H), 4.07-4.00 (m, 1H), 2.40 (s, 3H), 2.24-2.16 (m, 2H). 35 MS (ESI+): m/z 214 [M + H]. 1H NMR (300 M Hz, CD3OD) δ 8.26 (d, J = 8.7 Hz, 1H), 7.95 (t, J = 6.6 Hz, 2H), 7.70 (t, J = 7.2 Hz, 1H), 7.59 (t, J = 7.8 Hz, 1H), 7.35 (d, J = 8.7 Hz, 1H), 5.73-5.70 (m, 1H), 5.21 (d, J = 16.5 Hz, 1H), 5.05 (d, J = 16.2 Hz, 1H), 4.27-4.07 (m, 2H), 2.63-2.57 (m, 2H). 36 MS (ESI+): m/z 178 [M + H]. 1H NMR (300 M Hz, CD3OD) δ 7.21 (s, 2H), 7.12 (s, 1H), 4.85-4.72 (m, 3H), 4.20-4.12 (m, 1H), 4.07-3.99 (m, 1H), 2.35 (s, 3H), 2.32-2.20 (m, 2H). 37 MS (ESI+): m/z 178 [M + H]. 1H NMR (300 M Hz, CD3OD) δ 7.22-7.13 (m, 3H), 5.12 (d, J = 15 Hz, 1H), 4.67-4.62 (m, 2H), 4.16-4.07 (m, 1H), 3.99-3.90 (m, 1H), 2.30-2.25 (m, 5H). 38 MS (ESI+): m/z 198 [M + H]. 1H NMR (300 M Hz, DMSO- d6): δ 8.85 (brs, 3H), 7.48-7.43 (m, 2H), 7.34-7.30 (m, 1H), 4.78-4.65 (m, 3H), 4.11-4.06 (m, 1H), 3.97-3.89 (m, 1H), 2.13-1.91 (m, 2H). 39 MS (ESI+): m/z 178 [M + H]. 1H NMR (300 M Hz, CDCl3) δ 8.79 (brs, 3H), 7.28-7.13 (m, 2H), 6.95-7.00 (m, 1H), 5.12- 5.00 (m, 2H), 4.70 (d, J = 15.3 Hz, 1H), 4.31-4.21 (m, 1H), 4.01-3.95 (m, 1H), 2.70-2.51 (m, 4H), 2.31-2.20 (m, 1H). 40 MS (ESI+): m/z 198 [M + H]. 1H NMR (300 M Hz, DMSO- d6) δ 8.84 (brs, 3H), 7.46 (d, J = 6.0 Hz, 1H), 7.40 (d, J = 5.7 Hz, 1H), 7.33 (d, J = 5.7 Hz, 1H), 5.17 (d, J = 11.1 Hz, 1H), 4.85 (d, J = 5.7 Hz, 1H), 4.74 (d, J = 11.1 Hz, 1H), 4.11- 4.06 (m, 1H), 3.99-3.93 (m, 1H), 2.25-2.15 (m, 1H) 2.10- 2.00 (m, 1H). 41 MS (ESI+): m/z 198 [M + H]. 1H NMR (300 M Hz, CD3OD) δ 7.47 (d, J = 8.1 Hz, 1H), 7.36 (t, J = 7.5 Hz, 1H), 7.22 (d, J = 7.8 Hz, 1H), 5.26 (d, J = 5.4 Hz, 1H), 5.01 (d, J = 16.2 Hz, 1H), 4.87 (d, J = 16.2 Hz, 1H), 4.15-3.98 (m, 2H), 2.52- 2.37 (m, 2H).

Example 42: 5,6-Dimethylisochroman-4-amine hydrochloride

The compound of Example 42 was prepared as previously described in Example 21, using (2-bromo-3,4-dimethylphenyl)methanol (prepared from methyl 2-amino-3,4-dimethylbenzoate as previously described in Example 19). MS (ESI): m/z 178 [M+H]. 1H NMR (300 MHz, DMSO-d6) δ 8.32 (s, 3H), 7.20 (d, J=7.8 Hz, 1H), 6.90 (d, J=7.8 Hz, 1H), 4.83 (d, J=15.3 Hz, 1H), 4.68 (d, J=15.3 Hz, 1H), 4.48 (s, 1H), 4.24 (d, J=12.8 Hz, 1H), 3.76 (d, J=11.1 Hz, 1H), 2.25 (s, 6H).

Example 43: 6,7-Dimethyl-1,3,4,5-tetrahydrobenzo [c]oxepin-5-amine hydrochloride

The compound of Example 43 was prepared as previously described in Example 42 using but-3-en-1-ol. MS (ESI): m/z 192 [M+H]. 1H NMR (300 MHz, DMSO-d6) δ 8.44 (brs, 3H), 7.13 (d, J=7.5 Hz, 1H), 6.97 (d, J=7.8 Hz, 1H), 5.00 (d, J=15.0 Hz, 1H), 4.94 (s, 1H), 4.60 (d, J=15.0 Hz, 1H), 4.11-4.01 (m, 1H), 3.95-3.90 (m, 1H), 2.33 (s, 3H), 2.27 (s, 3H), 2.19-2.09 (m, 2H).

Example 44: 5,6-Dichloroisochroman-4-amine hydrochloride

The compound of Example 44 was prepared as previously described in Example 10, using (2-bromo-3,4-dichlorophenyl)methanol 44.5 prepared as described below. MS (ESI): m/z 218 [M+H]. 1H NMR (300 MHz, DMSO-d6) δ 8.62 (s, 3H), 7.72 (d, J=7.8 Hz, 1H), 7.26 (d, J=8.4 Hz, 1H), 4.90 (d, J=16.2 Hz, 1H), 4.69 (d, J=16.2 Hz, 1H), 4.52 (s, 1H), 4.31 (d, J=12.6 Hz, 1H), 3.82 (d, J=14.7 Hz, 1H).

Synthesis of (2-bromo-3,4-dichlorophenyl)methanol (44.5)

Step 1.

2-Amino-3,4-dichlorobenzoic acid 44.1 (13.6 g, 66.0 mmol, J. Med. Chem. 1991, 34, 218) was dissolved in DMF (150 mL) and treated with K2CO3 (45.6 g, 330 mmol) and iodomethane (11.2 g, 79.2 mmol). The resulting slurry was stirred at room temperature for 2 hr. The mixture was diluted with water and extracted with EtOAc (3×100 mL). The combined organics were washed with brine, dried over anhydrous Na2SO4, filtered, and concentrated under reduced pressure. Purification by flash column chromatography (SiO2, gradient elution from 100% hexanes to 10% EtOAc/hexanes) afforded methyl 2-amino-3,4-dichlorobenzoate 44.2 (12.5 g, 86%) as a yellow solid. 1H NMR (300 MHz, CDCl3) δ 7.74 (d, J=8.7 Hz, 1H), 6.74 (d, J=8.7 Hz, 1H), 6.47 (br s, 2H), 3.89 (s, 3H).

Step 2.

Methyl 2-bromo-3,4-dichlorobenzoate 44.3 was prepared as previously described in Example 19. 1H NMR (300 MHz, CDCl3) δ 7.54 (d, J=9.0 Hz, 1H), 7.49 (d, J=9.0 Hz, 1H), 3.94 (s, 3H).

Step 3.

Compound 44.3 (11.1 g, 39.0 mmol) was dissolved in MeOH (250 mL) and treated with aqueous 2 NNaOH (39.2 mL, 78 mmol), and stirred at room temperature for 2 hr. The mixture was filtered, diluted with water (800 mL), and made acidic to pH=2 by the addition of aqueous 3M HCl. The resulting suspension was extracted with EtOAc (4×400 mL). The combined organics were dried over anhydrous Na2SO4, filtered, and concentrated under reduced pressure to yield 2-bromo-3,4-dichlorobenzoic acid 44.4 (9.85 g, 94%), which was used directly without further purification.

Step 4.

2-Bromo-3,4-dichlorobenzyl alcohol 44.5 was prepared as previously described in Example 31. 1H NMR (300 MHz, CDCl3) δ 7.47 (d, J=8.4 Hz, 1H), 7.39 (d, J=8.4 Hz, 1H), 4.75 (s, 2H).

Example 45: 5-Chloro-6-methylisochroman-4-amine hydrochloride

The compound of Example 45 was prepared as previously described in Example 44 from 2-amino-3-chloro-4-methylbenzoic acid (Rewcastle, G. W., et al J. Med. Chem. 1991, 34, 217). MS (ESI): m/z 170 [M+H]. 1H NMR (300 MHz, CD3OD) δ 7.05 (d, J=0.9 Hz, 1H), 5.00 (d, J=15.3 Hz, 1H), 4.80 (d, J=15.3 Hz, 1H), 4.33 (s, 1H), 4.25 (dd, J=12.9, 1.5 Hz, 1H), 3.84 (dd, J=12.6, 1.8 Hz, 1H), 2.28 (d, J=1.2 Hz, 3H).

Example 46: 6,8-Dimethylisochroman-4-amine hydrochloride

The compound of Example 46 was prepared as previously described in Example 44 from 2-amino-4,6-dimethylbenzoic acid. MS (ESI): m/z 178 [M+H]. 1H NMR (300 MHz, CD3OD) δ 7.07 (s, 2H), 4.90 (d, J=18 Hz, 1H), 4.65 (d, J=18 Hz, 1H), 4.27 (s, 1H), 4.18 (dd, J=12, 3 Hz, 1H), 3.90 (dd, J=12, 3 Hz, 1H), 2.32 (s, 3H), 2.15 (s, 1H).

Example 47: 5,8-Dimethylisochroman-4-amine hydrochloride

The compound of Example 47 was prepared as previously described in Example 44 from 2-amino-3,6-dimethylbenzoic acid. MS (ESI): m/z 178 [M+H]. 1H NMR (300 MHz, DMSO-d6) δ 8.38 (s, 3H), 7.13-7.05 (m, 2H), 4.82 (d, J=15 Hz, 1H), 4.62 (d, J=15 Hz, 1H), 4.39 (s, 1H), 4.25 (d, J=12 Hz, 1H), 3.74 (d, J=12 Hz, 1H), 2.37 (s, 3H), 2.09 (s, 3H).

Example 48: 8-Chloro-5-methylisochroman-4-amine hydrochloride

The compound of Example 48 was prepared as previously described in Example 10, using (2-bromo-6-chloro-3-methylphenyl)methanol 48.5 prepared as described below. MS (ESI): m/z 198 [M+H]. 1H NMR (300 MHz, DMSO-d6) δ 8.46 (s, 3H), 7.42 (d, J=9.0 Hz, 1H), 7.23 (d, J=9.0 Hz, 1H), 4.85 (d, J=15 Hz, 1H), 4.65 (d, J=15 Hz, 1H), 4.46 (s, 1H), 4.25 (d, J=12 Hz, 1H), 3.79 (d, J=12 Hz, 1H), 2.41 (s, 3H).

Synthesis of (2-bromo-6-chloro-3-methylphenyl)methanol (48.5)

Step 1.

A solution of 5-chloro-2-methylaniline (15 g, 105 mmol) in water (750 mL) was treated with aqueous concentrated HCl (50 mL), hydroxylamine hydrochloride (36.4 g, 525 mmol), 2,2,2-trichloroethane-1,1-diol (25.9 g, 157 mmol) and Na2SO4 (14.9 g, 105 mmol), and heated to reflux for 2 h. After cooling to room temperature, the precipitate was collected by filtration, washed with water, and dried under vacuum to provide (E)-N-(5-chloro-2-methylphenyl)-2-(hydroxyimino)acetamide 48.1 (61.0 g) as a brown solid, which was used without further purification.

Step 2.

Compound 48.1 (51 g, 239 mmol) was dissolved in methanesulfonic acid (350 mL) and heating at 50° C. for 2 h. After cooling to room temperature, the mixture was poured into ice-water. The resulting suspension was collected on a fritted filter, washed with water, and dried under vacuum to provide 4-chloro-7-methylindoline-2,3-dione 48.2 (45.0 g, 96%) as a brown solid. 1H NMR (300 MHz, DMSO-d6) δ 10.22 (brs, 1H), 7.01 (dd, J=7.8, 0.6 Hz, 1H), 6.58 (d, J=8.1 Hz, 1H), 2.07 (s, 3H).

Step 3.

Compound 48.2 (15 g, 76.6 mmol) was dissolved in aqueous 0.3M NaOH (1.5 L) and treated with the dropwise addition of 40% hydrogen peroxide (150 mL). The reaction was stirred at room temperature for 4 h. Residual solids were removed by filtration. The filtrate was wasted with CH2Cl2 (3×200 mL), acidified to pH=3-4 with aqueous concentrated HCl, and extracted with EtOAc (3×350 mL). The combined organics were dried over anhydrous Na2SO4, filtered, and concentrated under reduced pressure to yield 2-amino-6-chloro-3-methylbenzoic acid 48.3 (12.7 g, 90%) as a yellow solid. 1H NMR (300 MHz, DMSO-d6) δ 7.02 (d, J=7.8 Hz, 1H), 6.58 (d, J=7.8 Hz, 1H), 2.07 (s, 3H).

Step 4.

Compound 48.3 (14.0 g, 75.4 mmol) was dissolved in EtOH (300 mL) and 48% hydrobromic acid (75 mL) and cooled in an ice bath. A solution of NaNO2 (7.79 g, 113 mmol) in water (20 mL) was added in a dropwise manner, and the resulting mixture was stirred at 0° C. for 20 min. Copper(I) bromide (10.8 g, 75.4 mmole) was added in a single portion, the ice bath was removed, and the suspension was heated with stirring to 95° C. for 30 min. After cooling to room temperature, saturated aqueous NH4Cl (200 mL) was added with vigorous stirring. The aqueous phase was extracted with EtOAc (3×200 mL). The combined organics were concentrated under reduced pressure. The residue was dissolved in water (30 mL) and made basic by the addition of aqueous 2N NaOH until pH=10. The aqueous phase was washed with CH2Cl2 (3×200 mL), made acidic to pH=3 by the addition of aqueous 5N HCl, and extracted with EtOAc (4×300 mL). The combined organics were dried over anhydrous Na2SO4, filtered, and concentrated under reduced pressure to yield 2-bromo-6-chloro-3-methylbenzoic acid 48.4. 1H NMR (300 MHz, DMSO-d6) δ 7.40-7.49 (m, 2H), 2.36 (s, 3H).

Step 5.

(2-Bromo-6-chloro-3-methylphenyl)methanol 48.5 was prepared as described previously in Example 44. 1H NMR (300 MHz, DMSO-d6) δ 7.39 (d, J=8.4 Hz, 1H), 7.32 (d, J=8.1 Hz, 1H), 4.75 (s, 2H), 2.36 (s, 3H).

Example 49: 8-Chloro-6-methylisochroman-4-amine hydrochloride

The compound of Example 49 was prepared as previously described in Example 48 from 3-chloro-5-methylaniline. MS (ESI): m/z 198 [M+H]. 1H NMR (300 MHz, CD3OD) δ 7.32 (s, 1H), 7.24 (s, 1H), 4.95 (d, J=15 Hz, 1H), 4.64 (d, J=15 Hz, 1H), 4.33 (s, 1H), 4.21 (dd, J=12.0, 3.0 Hz, 1H), 3.92 (dd, J=12.0, 3.0 Hz, 1H), 2.37 (s, 3H).

Example 50: 3,4-Dihydro-1H-benzo[h]isochromen-4-amine hydrochloride

The compound of Example 50 was prepared as previously described in Example 21 from 2-bromo-1-naphthalenemethanol (Shaik, F. H. et al, Beilstein J. Org. Chem. 2009, 5). MS (ESI): m/z 200 [M+H]. 1H NMR (300 MHz, DMSO-d6) δ 8.71 (d, J=6.6 Hz, 3H), 7.99-7.85 (m, 3H), 7.66-7.59 (m, 3H), 5.38 (d, J=15.9 Hz, 1H), 5.10 (d, J=15.9 Hz, 1H), 4.46 (s, 1H), 4.30 (d, J=12.6 Hz, 1H), 4.00 (dd, J=12.3, 2.1 Hz, 1H).

Example 51: 2,4,7,8,9,10-Hexahydro-1H-7,10-methanobenzo[f]isochromen-1-amine hydrochloride

The compound of Example 51 was prepared as previously described in Example 48 using 1,2,3,4-tetrahydro-1,4-methanonaphthalen-5-amine (Ehrenfreund, J.; et al, WO2004035589). MS (ESI): m/z 216 [M+H]. 1H NMR (300 MHz, CD3OD) δ 7.22 (dd, J=7.5, 2.4 Hz, 1H), 6.88 (d, J=7.5 Hz, 1H), 4.97-4.77 (m, 2H), 4.50 (d, J=41.1 Hz, 1H), 4.36-4.24 (m, 1H), 3.91 (d, J=12.9 Hz, 1H), 3.64 (d, J=12.0 Hz, 1H), 3.40-3.33 (m, 1H), 2.06-1.97 (m, 2H), 1.85-1.65 (m, 1H), 1.65-1.62 (m, 1H), 1.19-1.09 (m, 2H).

Example 52: 1,2,4,8,9,10-Hexahydropyrano[4,3-f]chromen-1-amine hydrochloride

The compound of Example 52 was prepared as previously described in Example 21 using (5-bromochroman-6-yl)methanol 52.5 prepared as described below. MS (ESI): m/z 206 [M+H]. 1H NMR (300 MHz, DMSO-d6) δ 8.40 (s, 3H), 6.88 (d, J=8.4 Hz, 1H), 6.80 (d, J=8.4 Hz, 1H), 4.79 (d, J=15.3 Hz, 1H), 4.63 (d, J=15.3 Hz, 1H), 4.34 (s, 1H), 4.22 (d, J=12.6 Hz, 1H), 4.16-4.06 (m, 2H), 3.74 (d, J=12.0 Hz, 1H), 3.00-2.90 (m, 1H), 2.78-2.68 (m, 1H), 1.98-1.89 (m, 2H).

Synthesis of (5-bromochroman-6-yl)methanol (52.5)

Step 1.

8-(tert-Butyl)-6-methylchroman 52.1 (3.5 g, 17.1 mmol, Mao, C.-H.; et al, Bioorg. Med. Chem. 2008, 16, 488) was dissolved in CH3CN (50 mL) and treated with N-bromosuccinimide (3.04 g, 17.1 mmol). The mixture was heated to 80° C. with stirring for 4 hr. After cooling to room temperature, the solvents were removed under reduced pressure, and the residue was purified by flash column chromatography (SiO2, 100% hexanes) to afford 5-bromo-8-(tert-butyl)-6-methylchroman 52.2 (2.5 g, 52%). 1H NMR (300 MHz, CDCl3) δ 7.03 (s, 1H), 4.13 (t, J=5.1 Hz, 2H), 2.81 (t, J=6.6 Hz, 2H), 2.37 (s, 3H), 2.08-2.00 (m, 2H), 1.41 (s, 9H).

Step 2.

AlCl3 (3.65 g, 27.4 mmol) was added to a solution of 52.2 (6.5 g, 22.9 mmol) in CH2Cl2 (100 mL) at room temperature and stirred for 2 h. The reaction mixture was carefully quenched by the dropwise addition of 1 M aqueous HCl (30 mL). The mixture was extracted with CH2Cl2 (200 mL). The organic phase was washed with brine (2×50 mL dried over anhydrous Na2SO4, filtered, and concentrated under reduced pressure. The resulting oil was purified by flash column chromatography (SiO2, 100% hexanes) to afford 5-bromo-6-methylchroman 52.3 (4.50 g, 86%) as a green oil. 1H NMR (300 MHz, CDCl3) δ 7.04 (d, J=8.4 Hz, 1H), 6.78 (d, J=8.4 Hz, 1H), 4.16 (t, J=5.1 Hz, 2H), 2.83 (t, J=6.6 Hz, 2H), 2.42 (s, 3H), 2.09-2.03 (m, 2H).

Step 3.

Chromium(VI) oxide (9.89 g, 99.0 mmol) was added to a solution of 52.3 (4.5 g, 19.8 mmol) in AcOH (40 mL) and water (20 mL). The reaction was stirred at ambient temperature for 2 h, diluted with water (40 mL), and extracted with EtOAc (200 mL). The organic phase was washed with saturated aqueous NaHCO3 (2×30 mL) and brine (2×30 mL), dried over anhydrous Na2SO4, filtered, and concentrated under reduced pressure. Purification by flash column chromatography (SiO2, 5% EtOAc/hexanes) gave 5-bromochroman-6-carbaldehyde 52.4 (1 g, 21%) as a white solid. 1H NMR (300 MHz, CDCl3) δ 10.31 (s, 1H), 7.75 (d, J=8.7 Hz, 1H), 6.87 (d, J=8.7 Hz, 1H), 4.24 (t, J=5.1 Hz, 2H), 2.85 (t, J=6.6 Hz, 2H), 2.13-2.06 (m, 2H).

Step 4.

NaBH4 (376 mg, 9.94 mmol) was added in a single portion to a solution of 52.4 (1.2 g, 4.97 mmol) in CH3OH (30 mL). The reaction mixture was stirred at room temperature for 30 min and then partitioned between water (30 mL) and EtOAc (300 mL). The organic phase was washed with brine (2×40 mL), dried over anhydrous Na2SO4, filtered, and concentrated under reduced pressure to provide (5-bromochroman-6-yl)methanol 52.5 (1.10 g, 91%) as a colorless oil which was used directly without further purification.

Example 53: 5-Ethylisochroman-4-amine hydrochloride

The compound of Example 53 was prepared as previously described in Example 21 using (2-bromo-3-ethylphenyl)methanol 53.4 prepared as described below. MS (ESI): m/z 178 [M+H]. 1H NMR (300 MHz, CD3OD) δ 7.36 (t, J=6.0 Hz, 1H), 7.24 (d, J=9.0 Hz, 1H), 7.00 (d, J=6.0 Hz, 1H), 4.95 (d, J=15.0 Hz, 1H), 4.84 (d, J=15.0 Hz, 1H), 4.52 (s, 1H), 4.25 (dd, J=12.0, 3.0 Hz, 1H), 3.92 (dd, J=12.0, 3.0 Hz, 1H), 2.85-2.66 (m, 2H), 1.27 (t, J=6.0 Hz, 3H).

Synthesis of (2-Bromo-3-ethylphenyl)methanol (53.4)

Step 1.

Methyl 2-amino-3-vinylbenzoate 53.1 was prepared as previously described in Example 7 using methyl 2-amino-3-bromobenzoate and vinylboronic acid pinacol ester. 1H NMR (300 MHz, CDCl3) δ 7.84 (d, J=9.0 Hz, 1H), 7.38 (d, J=6.0 Hz, 1H), 6.79-6.62 (m, 2H), 5.97 (brs, 2H), 5.62 (dd, J=18.0, 3.0 Hz, 1H), 5.38 (dd, J=12.0, 3.0 Hz, 1H), 3.87 (s, 3H).

Step 2.

Methyl 2-amino-3-ethylbenzoate 53.2 was prepared as previously described in Example 9. 1H NMR (300 MHz, CDCl3) δ 7.77 (d, J=9.0 Hz, 1H), 7.20 (d, J=6.0 Hz, 1H), 6.63 (t, J=6.0 Hz, 1H), 5.89 (brs, 2H), 3.86 (s, 1H), 2.54-2.47 (m, 2H), 1.26 (t, J=7.62 Hz, 3H).

Step 3.

Methyl 2-bromo-3-ethylbenzoate 53.3 was prepared as previously described in Example 19. 1H NMR (300 MHz, CDCl3) δ 7.45-7.42 (m, 1H), 7.36-7.25 (m, 2H), 3.92 (s, 3H), 2.87-2.80 (m, 2H), 1.24 (t, J=7.5 Hz, 3H).

Step 4.

(2-Bromo-3-ethylphenyl)methanol 53.4 was prepared as previously described in Example 19. 1H NMR (300 MHz, CDCl3) δ 7.32-7.23 (m, 2H), 7.19-7.16 (m, 1H), 4.76 (s, 2H), 2.84-2.76 (m, 2H), 2.17 (brs, 1H), 1.23 (t, J=15.0 Hz, 3H).

Example 54: N-Methylisochroman-4-amine hydrochloride

Step 1.

Isochroman-4-one (200 mg, 1.35 mmol) was dissolved in EtOH (10 mL) and treated sequentially with 2M NH3 in MeOH (3.4 mol, 6.8 mmol), Ti(Oi-Pr)4 (0.38 g, 1.35 mmol) and NaCNBH3 (0.34 g, 5.4 mmol). The reaction mixture was heated with stirring to reflux for 6 h and then cooled to room temperature and stirred for an additional 16 h. The reaction mixture was partitioned between EtOAc (40 mL) and saturated aqueous Na2CO3 (30 mL). The aqueous layer was extracted with EtOAc (2×15 mL). The combined organics were dried over anhydrous Na2SO4, filtered, and concentrated under reduced pressure to afford N-methylisochroman-4-amine (100 mg, 45%) as a colorless oil, which was converted to the compound of Example 54 as previously described in Example 1. MS (ESI): m/z 164 [M+H]. 1H NMR (300 MHz, DMSO-d6) δ 9.37 (brs, 1H), 9.20 (brs, 1H), 7.57 (d, J=7.5 Hz, 1H), 7.45-7.32 (m, 2H), 7.21 (d, J=7.5 Hz, 1H), 4.87 (d, J=15.6 Hz, 1H), 4.73 (d, J=15.6 Hz, 1H), 4.42 (d, J=13.2 Hz, 1H), 4.30 (brs, 1H), 3.87 (dd, J1=12.9, 2.1 Hz, 1H), 2.56-2.50 (m, 3H).

TABLE 5 Compounds prepared as described in Example 54 using the appropriate starting materials. Example Structure Characterization Data 55 MS (ESI): m/z 220 [M + H]. 1H NMR (300 M Hz, DMSO- d6) δ 8.52 (s, 1H), 8.34 (s, 1H), 7.23 (d, J = 9.0 Hz, 1H), 6.93 (d, J = 9.0 Hz, 1H), 4.90 (d, J = 15.0 Hz, 1H), 4.73 (d, J = 15.0 Hz, 1H), 4.61 (d, J = 9.0 Hz, 1H), 4.43 (d, J = 12.0 Hz, 1H), 3.71 (d, J = 15.0 Hz, 1H), 3.64-3.62 (m, 1H), 2.26 (d, J = 6.0 Hz, 6H), 1.38-1.09 (m, 6H). 56 MS (ESI): m/z 222 [M + H]. 1H NMR (300 M Hz, DMSO- d6) δ 8.80 (d, J = 24.9 Hz, 2H), 7.23 (d, J = 7.5 Hz, 1H), 6.92 (d, J = 7.2 Hz, 1H), 5.32 (s, 1H), 4.88 (d, J = 15.6 Hz, 1H), 4.70 (d, J = 15.3 Hz, 1H), 4.62-4.61 (m, 2H), 4.50 (d, J = 12.6 Hz, 1H), 3.77-3.70 (m, 3H), 3.13-3.07 (m, 2H), 2.26 (d, J = 4.8 Hz, 6H).

Example 57. N,5,6-Trimethylisochroman-4-amine hydrochloride

Step 1.

tert-Butyl (5,6-dimethylisochroman-4-yl)carbamate (400 mg, 1.44 mmol, prepared as described in Example 42) was dissolved in anhydrous THF (4 mL) and treated with a 95% dispersion of sodium hydride in mineral oil (103 mg, 4.31 mmol) at room temperature. Once addition was complete, the reaction mixture was heated to 40° C. and stirred for 15 min. Iodomethane (817 mg, 5.76 mmol) was added and heating at 40° C. was continued overnight. After cooling to room temperature, the reaction mixture was partitioned between brine (15 mL) and EtOAc (10 mL). The aqueous phase was washed with EtOAc (2×10 mL). The combined organics were dried over anhydrous Na2SO4, filtered, and concentrated under reduced pressure. Purification by flash column chromatography (SiO2, gradient elution from 3% EtOAc/hexanes to 5% EtOAc/hexanes) gave tert-butyl (5,6-dimethylisochroman-4-yl) (methyl)carbamate 57.1 (400 mg, 1.37 mmol) as a yellow oil. MS (ESI): m/z 292 [M+H].

Step 2.

The compound of Example 57 was prepared as previously described in Example 1. MS (ESI): m/z 192 [M+H]. 1H NMR (300 MHz, DMSO-d6) δ 8.85 (brs, 2H), 7.23 (d, J=7.8 Hz, 1H), 6.92 (d, J=7.8 Hz, 1H), 4.87 (d, J=15.3 Hz, 1H), 4.70 (d, J=15.3 Hz, 1H), 4.47 (d, J=9.3 Hz, 2H), 3.76 (dd, J=13.2, 1.8 Hz, 1H), 2.62 (s, 3H), 2.26 (s, 6H).

TABLE 6 Compounds prepared as described in Example 57 using the appropriate Boc-protected intermediates and alkyl halide. Example Structure Characterization Data 58 MS (ESI): m/z 206 [M + H]. 1H NMR (300 M Hz, DMSO- d6) δ 8.81 (s, 1H), 8.50 (s, 1H), 7.23 (d, J = 9.0 Hz, 1H), 6.93 (d, J = 9.0 Hz, 1H), 4.88 (d, J = 15.0 Hz, 1H), 4.71 (d, J = 15.0 Hz, 1H), 4.51-4.43 (m, 2H), 3.74 (d, J = 12.0 Hz, 1H), 3.12-3.09 (m, 1H), 2.25 (d, J = 6.0 Hz, 6H), 1.26 (t, J = 9.0 Hz, 3H). 59 MS (ESI): m/z 192 [M + H]. 1H NMR (300 M Hz, CD3OD) δ 7.26-7.16 (m, 3H), 5.01 (d, J = 15.6 Hz, 1H), 4.83 (d, J = 15.3 Hz, 1H), 4.50-4.47 (m, 1H), 4.15-4.09 (m, 1H), 4.03-3.96 (m, 1H), 2.73 (s, 3H), 2.47-2.39 (m, 5H). 258 MS (ESI): m/z 192 [M + H]. 1H NMR (300 M Hz, DMSO- d6) δ 9.08 (s, 1H), 8.55 (s, 1H), 7.51 (d, J = 7.8 Hz, 1H), 7.45-7.42 (m, 1H), 7.33 (t, J = 7.2 Hz, 1H), 7.22 (d, J = 7.8 Hz, 1H), 4.97 (d, J = 15.9 Hz, 1H), 4.80 (d, J = 15.9 Hz, 1H), 4.32 (s, 1H), 3.75-3.70 (m, 1H), 2.42 (s, 3H), 1.80-1.70 (m, 2H), 1.02 (t, J = 7.2 Hz, 3H). 259 MS (ESI): m/z 192 [M + H]. 1H NMR (300 M Hz, DMSO- d6) δ 9.58 (s, 1H), 9.00 (s, 1H), 7.51 (d, J = 7.5 Hz, 1H), 7.43-7.40 (m, 1H), 7.35-7.30 (m, 1H), 7.21 (d, J = 7.5 Hz, 1H), 4.80 (d, J = 16.2 Hz, 1H), 4.69 (d, J = 16.5 Hz, 1H), 4.29-4.27 (m, 1H), 4.18 (s, 1H), 2.51 (s, 3H), 1.52-1.38 (m, 2H), 0.95 (t, J = 7.5 Hz, 3H). 262 MS (ESI): m/z 192 [M + H]. 1H NMR (300 M Hz, CD3OD) δ 7.48-7.43 (m, 2H), 7.39-7.34 (m, 1H), 7.21 (d, J = 7.5 Hz, 1H), 4.97 (d, J = 15.6 Hz, 1H), 4.81 (d, J = 15.6 Hz, 1H), 4.43 (dd, J = 13.2, 1.2 Hz, 1H), 4.29 (s, 1H), 3.92 (dd, J = 13.2, 1.8 Hz, 1H), 3.11-3.06 (m, 2H), 1.81-1.68 (m, 2H), 1.02 (t, J = 7.4 Hz, 3H). 263 MS (ESI): m/z 188 [M + H]. 1H NMR (300 M Hz, CD3OD) δ 7.47 (t, J = 7.5 Hz, 2H), 7.37 (t, J = 7.8 Hz, 1H), 7.22 (d, J = 7.5 Hz, 1H), 4.98 (d, J = 15.6 Hz, 1H), 4.83 (d, J = 15.6 Hz, 1H), 4.48-4.41 (m, 2H), 4.09-3.90 (m, 3H), 3.33 (t, J = 2.4 Hz, 1H). 264 MS (ESI): m/z 206 [M + H]. 1H NMR (300 M Hz, CDCl3) δ 7.42-7.39 (m, 2H), 7.32-7.27 (m, 1H), 7.15 (d, J = 7.8 Hz, 1H), 4.80-4.75 (m, 2H), 4.51-4.48 (m, 1H), 4.08 (s, 1H), 3.01-2.87 (m, 2H), 1.71-1.62 (m, 2H), 1.18 (d, J = 6.6 Hz, 3H), 0.94 (t, J = 7.5 Hz, 3H). 265 MS (ESI): m/z 192 [M + H]. 1H NMR (300 M Hz, CD3OD) δ 7.45 (t, J = 7.5 Hz, 2H), 7.37 (d, J = 7.8 Hz, 1H), 7.21 (d, J = 7.5 Hz, 1H), 4.97 (d, J = 15.6 Hz, 1H), 4.81 (d, J = 15.6 Hz, 1H), 4.43 (dd, J = 13.2, 1.2 Hz, 1H), 4.29 (s, 1H), 3.92 (dd, J = 13.2, 1.8 Hz, 1H), 3.13-3.06 (m, 2H), 1.81- 1.68 (m, 2H), 1.02 (t, J = 7.5 Hz, 3H). 266 MS (ESI): m/z 188 [M + H]. 1H NMR (300 M Hz, CD3OD) δ 7.47 (t, J = 7.2 Hz, 2H), 7.37 (t, J = 7.5 Hz, 1h), 7.22 (d, J = 7.5 Hz, 1H), 4.97 (d, J = 15.6 Hz, 1H), 4.82 (d, J = 15.6 Hz, 1H), 4.48-4.41 (m, 2H), 4.08-3.90 (m, 3H), 3.32 (t, J = 2.4 Hz, 1H). 267 MS (ESI): m/z 202 [M + H]. 1H NMR (300 M Hz, CDCl3) δ 7.62 (d, J = 7.5 Hz, 1H), 7.42-7.28 (m, 2H), 7.10 (d, J = 7.5 Hz, 1H), 5.05-5.00 (m, 2H), 4.78 (d, J = 15.9 Hz, 1H), 4.36 (s, 1H), 4.09 (dd, J = 17.1, 2.4 Hz, 1H), 3.50 (dd, J = 17.1, 2.4 Hz, 1H), 2.60 (t, J = 2.4 Hz, 1H), 1.26 (d, J = 6.6 Hz, 3H). 268 MS (ESI): m/z 206 [M + H]. 1H NMR (300 M Hz, CD3OD) δ 7.42-7.39 (m, 2H), 7.32-7.27 (m, 1H), 7.15 (d, J = 7.8 Hz, 1H), 4.80-4.75 (m, 2H), 4.51-4.48 (m, 1H), 4.08 (s, 1H), 3.01-2.87 (m, 2H), 1.71-1.62 (m, 2H), 1.18 (d, J = 6.6 Hz, 3H), 0.94 (t, J = 7.5 Hz, 3H). 269 MS (ESI): m/z 206 [M + H]. 1H NMR (300 M Hz, CD3OD) δ 7.27-7.21 (m, 2H), 7.16-7.11 (m, 1H), 7.01 (d, J = 7.8 Hz, 1H), 4.79 (d, J = 15.6 Hz, 1H), 4.65 (d, J = 15.6 Hz, 1H), 4.07 (s, 1H), 3.89-3.86 (m, 1H), 2.70 (t, J = 8.4 Hz, 2H), 1.61-1.41 (m, 2H), 1.26 (d, J = 6.9 Hz, 3H), 0.74 (t, J = 7.5 Hz, 3H). 270 MS (ESI): m/z 202 [M + H]. 1H NMR (300 M Hz, CDCl3) δ 7.62 (d, J = 7.2 Hz, 1H), 7.42-7.28 (m, 2H), 7.10 (d, J = 7.5 Hz, 1H), 5.05-5.00 (m, 2H), 4.78 (d, J = 15.9 Hz, 1H), 4.36 (s, 1H), 4.09 (dd, J = 17.1, 2.4 Hz, 1H), 3.50 (dd, J = 17.1, 2.4 Hz, 1H), 2.60 (t, J = 2.4 Hz, 1H), 1.26 (d, J = 6.6 Hz, 3H). 271 MS (ESI): m/z 202 [M + H]. 1H NMR (300 M Hz, CDCl3) δ 7.68 (d, J = 7.5 Hz, 1H), 7.41-7.26 (m, 2H), 7.09 (d, J = 7.5 Hz, 1H), 5.13 (d, J = 15.6 Hz, 1H), 4.85 (d, J = 15.6 Hz, 1H), 4.38 (s, 1H), 4.21 (dd, J = 16.8, 2.4 Hz, 1H), 4.06 (q, J = 6.9 Hz, 1H), 3.52 (dd, J = 16.8, 2.4 Hz, 1H), 2.57 (t, J = 2.4 Hz, 1H), 1.74 (d, J = 6.9 Hz, 3H). 272 MS (ESI): m/z 206 [M + H]. 1H NMR (300 M Hz, CDCl3) δ 10.09 (brs, 1H), 9.23 (brs, 1H), 8.20-8.17 (m, 1H), 7.40- 7.37 (m, 2H), 7.12-7.09 (m, 1H), 5.10 (d, J = 15.6 Hz, 1H), 4.82 (d, J = 15.6 Hz, 1H), 4.48 (s, 1H), 4.06-4.02 (m, 1H), 3.20-3.06 (m, 1H), 2.65-2.47 (m, 1H), 2.05-1.74 (m, 2H), 1.75 (d, J = 6.9 Hz, 3H), 0.88 (t, J = 7.2 Hz, 3H).

Example 60. N,N-dimethylisochroman-4-amine hydrochloride

Step 1.

The compound of Example 57 (200 mg, 1.04 mmol) was dissolved in CH3OH (4 mL). Paraformaldehyde (93.6 mg, 3.12 mmol), NaBH3CN (196 mg, 3.12 mmol), and acetic acid (187 mg, 3.12 mmol) were added, and the reaction was stirred at room temperature for 1 h. The reaction mixture was poured into saturated aqueous Na2CO3 (10 mL) and extracted with EtOAc (4×10 mL). The combined organics were dried over anhydrous Na2SO4, filtered, and concentrated under reduced pressure. Purification by flash column chromatography (SiO2, 100% EtOAc) gave N,N,-dimethylisochroman-4-amine (120 mg, 56%) as a white oil. The compound of Example 60 was prepared as described previously in Example 1 as an off-white solid. MS (ESI): m/z 178 [M+H]. 1H NMR (300 MHz, DMSO-d6) δ 10.65 (brs, 1H), 7.54 (d, J=7.5 Hz, 1H), 7.48-7.43 (m, 1H), 7.37 (t, J=7.5 Hz, 1H), 7.23 (d, J=7.8 Hz, 1H), 4.89 (d, J=15.6 Hz, 1H), 4.73-4.65 (m, 2H), 4.49 (s, 1H), 3.85 (dd, J=13.8, 2.7 Hz, 1H), 2.82 (d, J=4.8 Hz, 3H), 2.64 (d, J=4.5 Hz, 3H).

TABLE 7 Compounds prepared as described in Example 60 using the appropriate amine. Example Structure Characterization Data 61 MS (ESI): m/z 206 [M + H]. 1H NMR (300 M Hz, CD3OD) δ 7.31 (d, J = 8.1 Hz. 1H), 6.98 (d, J = 7.8 Hz, 1H), 4.97 (d, J = 15.6 Hz, 1H), 4.79 (d, J = 15.6 Hz, 1H), 4.67 (d, J = 12.6 Hz, 2H), 3.88 (d, J = 12.6 Hz, 1H), 2.92 (s, 6H), 2.35 (d, J = 2.7 Hz, 6H). 62 MS (ESI): m/z 206 [M + H]. 1H NMR (300 M Hz, CD3OD) δ 7.29-7.26 (m, 2H), 7.19 (d, J = 7.8 Hz, 1H), 5.14 (d, J = 15.9 Hz, 1H), 4.81 (d, J = 15.9 Hz, 1H), 4.45 (d, J = 3.6 Hz, 1H), 4.21-4.15 (m, 1H), 4.00-3.96 (m, 1H), 3.13 (s, 3H), 2.72-2.68 (m, 1H), 2.66 (s, 3H), 2.41-2.48 (m, 1H), 2.38 (s, 3H). 260 MS (ESI): m/z 206 [M + H]. 1H NMR (300 M Hz, DMSO-d6) δ 7.55-7.50 (m, 2H), 7.37 (t, J = 7.2 Hz, 1H), 7.27 (d, J = 7.5 Hz, 1H), 5.02 (d, J = 15.9 Hz, 1H), 4.79 (d, J = 15.9 Hz, 1H), 4.55 (s, 1H), 3.80- 372 (m, 1H), 2.86 (d, J = 5.1 Hz, 3H), 2.46 (d, J = 4.8 Hz, 3H), 1.97-1.87 (m, 2H), 1.03 (t, J = 7.2 Hz, 3H). 261 MS (ESI): m/z 206 [M + H]. 1H NMR (300 M Hz, DMSO-d6) δ 7.63 (d, J = 7.5 Hz, 1H), 7.49-7.44 (m, 1H), 7.36 (t, J = 7.2 Hz, 1H), 7.23 (d, J = 7.8 Hz, 1H), 4.83 (d, J = 16.5 Hz, 1H), 4.66 (d, J = 16.5 Hz, 1H), 4.54-49 (m, 1H), 4.38 (s, 1H), 2.70 (s, 6H), 1.50-1.39 (m, 2H), 0.96 (t, J = 7.2 Hz, 3H). 283 MS (ESI): m/z 279 [M + H]. 1H NMR (300 M Hz, CD3OD) δ 7.48 (d, J = 8.4 Hz, 1H), 7.06 (d, J = 8.4 Hz, 1H), 6.94 (s, 1H), 4.87 (d, J = 15.9 Hz, 1H), 4.68 (d, J = 15.9 Hz, 1H), 4.57 (d, J = 13.9 Hz, 1H), 4.31 (s, 1H), 3.86-3.79 (m, 1H), 3.44-3.26 (m, 2H), 2.95 (d, J = 33.9 Hz, 3H), 2.85 (s, 3H), 2.81 (s, 3H), 1.17 (dt, J = 22.5, 6.9 Hz, 3H). 284 MS (ESI): m/z 279 [M + H]. 1H NMR (300 M Hz, CD3OD) δ 7.25 (d, J = 1.8 Hz, 1H), 7.20-7.15 (m, 2H), 4.88 (d, J = 15.6 Hz, 1H), 4.68 (d, J = 15.6 Hz, 1H), 4.55 (d, J = 13.8 Hz, 1H), 4.27 (s, 1H), 3.82 (dd, J = 14.1, 2.4 Hz, 1H), 3.43-3.27 (m, 2H), 2.95 (d, J = 36.0 Hz, 3H), 2.82 (s, 6H), 1.20-1.06 (m, 3H). 285 MS (ESI): m/z 293 [M + H]. 1H NMR (300 M Hz, CD3OD) δ 7.34-7.27 (m, 3H), 5.05 (d, J = 15.9 Hz, 1H), 4.89 (d, J = 15.9 Hz, 1H), 4.48-4.45 (m, 1H), 4.20-4.11 (m, 1H), 3.50-3.36(m, 2H), 3.06 (d, J = 36.3 Hz, 3H), 3.05 (s, 3H), 2.70 (s, 3H), 1.58 (d, J = 6.9 Hz, 3H), 1.24 (dt, J = 25.1, 7.2 Hz, 3H). 286 MS (ESI): m/z 293 [M + H]. 1H NMR (300 M Hz, CD3OD) δ 7.34-7.26 (m, 3H), 4.90-4.88 (m, 2H), 4.83-4.76 (m, 1H), 4.26 (s, 1H), 3.58-3.36 (m, 2H), 3.06 (d, J = 36.0 Hz, 3H), 2.95 (s, 3H), 2.88 (s, 3H), 1.31-1.18 (m, 6H). 287 MS (ESI): m/z 293 [M + H]. 1H NMR (300 M Hz, CD3OD) δ 7.26-7.16 (m, 3H), 4.99 (d, J = 15.6 Hz, 1H), 4.85 (d, J = 15.6 Hz, 1H), 4.28 (s, 1H), 4.05 (q, J = 6.6 Hz, 1H), 3.57-3.31 (m, 2H), 3.05 (d, J = 34.1 Hz, 3H), 1.41 (d, J = 6.6 Hz, 3H), 1.24 (dt, J = 23.7, 7.2 Hz, 3H). 288 MS (ESI): m/z 293 [M + H]. 1H NMR (300 M Hz, CD3OD) δ 7.09-7.00 (m, 3H), 4.69 (s, 2H), 4.17- 4.04 (m, 2H), 3.40-3.32 (m, 2H), 2.89 (d, J = 34.8 Hz, 3H), 1.17 (d, J = 6.6 Hz, 3H), 1.09 (dt, J = 24.6, 7.2 Hz, 3H).

Example 63. 4,4,7-Trimethyl-1,3,4,5-tetrahydrobenzo[c]oxepin-5-amine hydrochloride

The compound of Example 63 was prepared as previously described in Example 1, using 4,4,7-trimethyl-3,4-dihydrobenzo[c]oxepin-5(1H)-one (prepared as described below). MS (ESI): m/z 206 [M+H]. 1H NMR (300 MHz, CD3OD) δ 7.24-7.16 (m, 3H), 4.93-4.76 (m, 2H), 4.29 (s, 1H), 3.80 (d, J=12.6 Hz, 1H), 3.61 (d, J=12.6 Hz, 1H), 2.39 (s, 3H), 1.06 (s, 3H), 0.97 (s, 3H).

Synthesis of 4,7-methyl-3,4-dihydrobenzo[c]oxepin-5(1H)-one and 4,4,7-trimethyl-3,4-dihydrobenzo[c]oxepin-5(1H)-one

Step 1.

A solution of 1.2 g (6.8 mmol) of 7-methyl-3,4-dihydrobenzo[c]oxepin-5(1H)-one (prepared as previously described in Example 34) in DMF (100 mL) was treated with iodomethane (9.63 g, 67.9 mmol) and cesium carbonate (11.0 g, 33.9 mmol). The reaction was heated with stirring at 50° C. for 2 h, and then partitioned between EtOAc (250 mL) and water (300 mL). The organic phase was washed with brine (100 mL) dried over anhydrous Na2SO4, filtered, and concentrated under reduced pressure. Purification by flash column chromatography (SiO2, gradient elution from 100% hexanes to 5% EtOAc/hexanes) gave 4,7-dimethyl-3,4-dihydrobenzo[c]oxepin-5(1H)-one (550 mg, 43%) as a colorless oil and 4,4,7-trimethyl-3,4-dihydrobenzo[c]oxepin-5(1H)-one (320 mg, 23%) as a colorless oil.

4,7-dimethyl-3,4-dihydrobenzo[c]oxepin-5(1H)-one: MS (ESI)

m/z 190 [M+H].

4,4,7-trimethyl-3,4-dihydrobenzo[c]oxepin-5(1H)-one

MS (ESI): m/z 204 [M+H].

Example 68. 6-Fluoro-4-methylisochroman-4-amine hydrochloride

Step 1.

A solution of 6-fluoroisochroman-4-one (420 mg, 2.52 mmol, prepared as described in Example 6) in diethyl ether (40 mL) was cooled in an ice bath and treated with methylmagnesium bromide (3.78 mL, 3.78 mmol, 1M in THF). The reaction was stirred at 0° C. for 1 h. Saturated aqueous NH4Cl (60 mL) and EtOAc (40 mL) were added to the reaction vessel and the resulting biphasic mixture was transferred to a separatory funnel. The layers were separated and the aqueous phase was washed with EtOAc (2×40 mL). The combined organics were dried over anhydrous Na2SO4, filtered, and concentrated under reduced pressure to provide 6-fluoro-4-methylisochroman-4-ol 68.1 (455 mg, 99%) as yellow oil. GC-MS (ESI) m/z 182 [M+H].

Step 2.

Compound 68.1 (250 mg, 1.37 mmol) was dissolved in toluene (6 mL) and cooled in an ice bath. Azidotrimethylsilane (394 mg, 3.42 mmol) and BF3-Et2O (1.24 g, 4.11 mmol) were added, the ice bath was removed, and the reaction was stirred at 30° C. for 2 days. The reaction mixture was carefully quenched by the slow addition of NH4OH and extracted with EtOAc (3×50 mL). The combined organic layers were washed with brine, dried over anhydrous Na2SO4, filtered, and concentrated under reduced pressure. Purification by flash column chromatography (SiO2, 100% hexanes) afforded 4-azido-6-fluoro-4-methyl isochroman 68.2 (240 mg, 84%) as yellow oil. 1HNMR (300 MHz, CDCl3) δ 7.19 (d, J=1.8 Hz, 1H), 7.05-6.99 (m, 2H), 4.86 (d, J=15.0 Hz, 1H), 4.74 (d, J=14.7 Hz, 1H), 4.05 (d, J=11.7 Hz, 1H), 3.78 (d, J=12.0 Hz, 1H), 1.49 (s, 3H).

Step 3.

Compound 68.2 (240 mg, 1.15 mmol) was dissolved in THF (10 mL), cooled in an ice bath, and treated with LiAlH4 (87.2 mg, 2.30 mmol) in a single portion. The ice bath was removed, and the reaction mixture was stirred at room temperature for 2 h. Excess hydride was quenched by the sequential addition of water (0.5 mL) and aqueous 2M NaOH (0.5 mL). The resulting mixture was partitioned between aqueous 1M HCl (40 mL) and CH2Cl2 (50 mL). The aqueous phase was washed with CH2Cl2 (2×50 mL) and then made basic by the addition of aqueous 1M NaOH (40 mL). The aqueous phase was extracted with CH2Cl2 (4×50 mL). The combined organics were dried over anhydrous Na2SO4, filtered, and concentrated under reduced pressure to give crude 6-fluoro-4-methylisochroman-4-amine, which was purified by preparative HPLC and converted to the compound of Example 68 (67 mg, 27%) as a white solid as described previously in Example 1. MS (ESI): m/z 182 [M+H]. 1H NMR (300 MHz, CD3OD) δ 7.39 (dd, J=9.9, 2.4 Hz, 1H), 7.24-7.15 (m, 2H), 4.94 (d, J=15.2 Hz, 1H), 4.83 (d, J=15.3 Hz, 1H), 4.06 (d, J=12.6 Hz, 1H), 3.82 (d, J=12.6 Hz, 1H), 1.66 (s, 3H).

Example 72. cis-3-methylisochroman-4-amine hydrochloride Example 73. trans-3-methylisochroman-4-amine hydrochloride

Step 1.

3-Methylisochroman-4-one 72.1 was prepared as previously described in Example 69 using isochroman-4-one (prepared as previously described in Example 12) and iodomethane. MS (ESI): m/z 163 [M+H]. 1HNMR (300 MHz, CDCl3) δ 8.05 (d, J=7.5 Hz, 1H), 7.59-7.53 (m, 1H), 7.49-7.39 (m, 1H), 7.21 (d, J=7.5 Hz, 1H), 5.18 (s, 2H), 4.28 (q, J=6.6 Hz, 1H), 1.75 (d, J=6.6 Hz, 3H).

Step 2.

3-Methylisochroman-4-ol 72.2 was prepared as previously described in Example 1. 1H NMR (300 MHz, CDCl3) δ 7.56-7.45 (m, 1H), 7.34-7.24 (m, 2H), 7.07-7.01 (m, 1H), 4.89-4.82 (m, 2H), 4.46-4.40 (m, 0.5H), 4.40-4.25 (m, 0.5H), 4.15-4.13 (m, 0.5H), 3.69-3.64 (m, 0.5H), 1.44 (d, J=6.3 Hz, 3H).

Step 3.

4-Azido-3-methylisochroman 72.3 was prepared as previously described in Example 1. 1H NMR (300 MHz, CDCl3) δ 7.45 (d, J=8.7 Hz, 1H), 7.43-7.24 (m, 2H), 7.02 (d, J=8.4 Hz, 1H), 4.80 (d, J=2.7 Hz, 2H), 4.11 (d, J=7.8 Hz, 1H), 3.80-3.76 (m, 1H), 1.44 (d, J=6.3 Hz, 3H).

Step 4.

LiAlH4 (119 mg, 3.15 mmol) was added to a room temperature solution of compound 72.3 (200 mg, 1.05 mmol) in THF (10 mL) in a single portion, and the reaction was stirred for 1 h. A solution of aqueous 1M NaOH (5 mL), di-tert-butyl dicarbonate (916 mg, 4.20 mmol) and Na2CO3 (333 mg, 3.15 mmol) were added to the reaction vessel, and the biphasic mixture was stirred vigorously for 2 h. After transferring to a separatory funnel, the mixture was partitioned between water (20 mL) and EtOAc (100 mL). The organic phase was washed with brine (40 mL), dried over anhydrous Na2SO4, filtered, and concentrated under reduced pressure. Purification by flash column chromatography (SiO2, gradient elution from 100% hexanes to 5% EtOAc/hexanes) provided tert-butyl ((3S,4S)-3-methylisochroman-4-yl) carbamate 72.4a (90.0 mg, 32%) as a white solid and tert-butyl ((3R,4S)-3-methylisochroman-4-yl)carbamate 72.4b (70.0 mg, 25%) as a white solid.

Rac-tert-butyl ((3 S,4S)-3-methylisochroman-4-yl)carbamate 72.4a TLC higher Rf. MS (ESI): m/z 286 [M+Na]. 1HNMR (300 MHz, CDCl3) δ 7.457.42 (m, 1H), 7.28-7.25 (m, 2H), 7.03-7.00 (m, 1H), 4.954.81 (m, 1H), 4.82 (s, 2H), 4.64 (d, J=8.7 Hz, 1H), 3.91-3.84 (m, 1H), 1.46 (s, 9H), 1.34 (d, J=6.3 Hz, 3H).
Rac-tert-butyl ((3R,4S)-3-methylisochroman-4-yl)carbamate 72.4b TLC lower Rf. MS (ESI): m/z 286 [M+Na]. 1HNMR (300 MHz, CDCl3) δ 7.42-7.37 (m, 1H), 7.28-7.21 (m, 2H), 7.02-7.00 (m, 1H), 4.81 (s, 2H), 4.79-4.65 (m, 2H), 3.72-3.67 (m, 1H), 1.48 (s, 9H), 1.38 (d, J=6.3 Hz, 3H).

Step 5.

The compounds of Example 72 and Example 73 were prepared as previously described in Example 1.

Example 72

MS (ESI): m/z 164 [M+H]. 1H NMR (300 MHz, CDCl3) δ 7.42-7.33 (m, 3H), 7.18 (d, J=6.9 Hz, 1H), 4.99-4.83 (m, 2H), 4.25 (s, 1H), 4.03-4.05 (m, 1H), 1.40 (d, J=6.6 Hz, 3H).

Example 73

MS (ESI): m/z 164 [M+H]. 1H NMR (300 MHz, CD3OD) δ 7.46-7.37 (m, 3H), 7.21 (d, J=7.2 Hz, 1H), 4.97-4.78 (m, 2H), 4.37-4.20 (m, 2H), 1.33 (d, J=6.6 Hz, 3H).

Example 74. cis-4,7-dimethyl-1,3,4,5-tetrahydrobenzo [c]oxepin-5-amine hydrochloride

The compound of Example 74 was prepared as previously described in Example 72, using 4,7-methyl-3,4-dihydrobenzo[c]oxepin-5(1H)-one (prepared as described in Example 63). MS (ESI): m/z 192 [M+H]. 1H NMR (300 MHz, CD3OD) δ 7.23-7.06 (m, 3H), 5.07 (d, J=15.3 Hz, 1H), 4.79 (d, J=15.3 Hz, 1H), 4.28 (s, H), 4.21-4.15 (m, 1H), 3.61-3.54 (m, 1H), 2.52-2.44 (m, 1H), 2.38 (s, 3H), 0.93 (d, J=7.2 Hz, 3H).

Example 75. trans-4,7-dimethyl-1,3,4,5-tetrahydrobenzo [c]oxepin-5-amine hydrochloride

The compound of Example 75 was prepared as previously described in Example 72, using 4,7-methyl-3,4-dihydrobenzo[c]oxepin-5(1H)-one (prepared as described in Example 63). MS (ESI): m/z 192 [M+H]. 1H NMR (300 MHz, CD3OD) δ 7.20-7.17 (m, 2H), 7.11 (s, 1H), 4.82-4.69 (m, 3H), 4.10-4.05 (m, 1H), 3.88-3.82 (m, 1H), 2.50-2.41 (m, 1H), 2.40 (s, 3H). 0.94 (d, J=6.9 Hz, 3H).

Example 76. cis-3,6-Dimethylisochroman-4-amine hydrochloride Example 77. trans-3,6-Dimethylisochroman-4-amine hydrochloride

Step 1.

2-Bromo-1-((but-3-en-2-yloxy)methyl)-4-methylbenzene 76.1 was prepared as previously described in Example 10 using 2-bromo-1-(bromomethyl)-4-methylbenzene and but-3-en-2-ol. MS (ESI): m/z 256 [M+H]. 1H NMR (300 MHz, CDCl3) δ 7.38 (s, 1H), 7.37 (d, J=7.8 Hz, 1H), 7.12 (d, J=7.8 Hz, 1H), 5.89-5.78 (m, 1H), 5.29-5.18 (m, 2H), 4.57 (d, J=12.9 Hz, 1H), 4.45 (d, J=12.9 Hz, 1H), 2.33 (s, 3H), 1.33 (d, J=6.3 Hz, 3H).

Step 2.

3,6-Dimethyl-4-methyleneisochroman 76.2 was prepared as previously described in Example 10. MS (ESI): m/z 175 [M+H]. 1H NMR (300 MHz, CDCl3) δ 7.49 (s, 1H), 7.08 (d, J=7.8 Hz, 1H), 6.95 (d, J=7.8 Hz, 1H), 5.63 (d, J=0.9 Hz, 1H), 5.07 (d, J=1.5 Hz, 1H), 4.82 (d, J=6.9 Hz, 2H), 4.45-4.36 (m, 1H), 2.37 (s, 3H), 1.51 (d, J=6.6 Hz, 3H).

Step 3.

3,6-Dimethylisochroman-4-one 76.3 was prepared as previously described in Example 10. MS (ESI): m/z 177 [M+H]. 1H NMR (300 MHz, CDCl3) δ 7.86 (s, 1H), 7.37 (d, J=7.5 Hz, 1H), 7.11 (d, J=7.5 Hz, 1H), 4.91 (s, 2H), 4.25 (q, J=6.6 Hz, 1H), 2.41 (s, 3H), 1.50 (d, J=6.6 Hz, 3H).

Step 4.

3,6-Dimethylisochroman-4-ol 76.4 was prepared as previously described in Example 1. 1H NMR (300 MHz, CDCl3) δ 7.30-7.24 (m, 1H), 7.14-7.06 (m, 1H), 6.95-6.90 (m, 1H), 4.78-4.76 (m, 2H), 4.01-4.35 (m, 0.75H), 4.25-4.21 (m, 0.25H), 3.80-3.77 (m, 0.25H), 3.68-3.60 (m, 0.75H), 2.37 (s, 3H), 1.42 (d, J=6.3 Hz, 3H).

Step 5.

4-Azido-3,3-dimethylisochroman 76.5 was prepared as previously described in Example 1.

Step 6.

Compound 76.5 (500 mg, 2.46 mmol) was dissolved in THF (30 mL) and treated with PPh3 (645 mg, 2.46 mmol). The reaction was stirred at room temperature for 30 min. Water (10 mL) was added, and the solution was heated to 60° C. for 12 h. After cooling to room temperature, the mixture was partitioned between aqueous 1M HCl (30 mL) and EtOAc (50 mL). The aqueous phase was washed with EtOAc (2×30 mL). The aqueous phase was made basic by the addition of Na2CO3 (521 mg, 4.92 mmol), and then treated with di-tert-butyl dicarbonate (1.07 g, 4.92 mmol) and THF (40 mL). The biphasic mixture was stirred vigorously at room temperature for 30 min, and then partitioned between water (30 mL) and EtOAc (50 mL). The aqueous phase was extracted with EtOAc (2×50 mL). The combined organics were washed with brine, dried over anhydrous Na2SO4, filtered, and concentrated under reduced pressure. Purification by flash column chromatography (SiO2, gradient elution from 100% hexanes to 97% EtOAc/hexanes) gave tert-butyl-(cis-3,6-dimethylisochroman-4-yl)carbamate 76.5a (300 mg, 44%) as a white solid and tert-butyl-(trans-3,6-dimethylisochroman-4-yl)carbamate 76.5b (170 mg, 25%) as a white solid.

tert-butyl-(cis-3,6-dimethylisochroman-4-yl)carbamate 76.5a TLC higher Rf. MS (ESI): m/z 300 [M+Na].

tert-butyl-(trans-3,6-dimethylisochroman-4-yl)carbamate 76.5b TLC lower Rf. MS (ESI): m/z 300 [M+Na].

Step 7.

The compounds of Example 76 and Example 77 were prepared as previously described in Example 1.

Example 76

MS (ESI): m/z 178 [M+H]. 1H NMR (300 MHz, DMSO-d6) δ 8.27 (brs, 3H), 7.31 (s, 1H), 7.21 (d, J=7.8 Hz, 1H), 7.06 (d, J=7.8 Hz, 1H), 4.86-4.71 (m, 2H), 4.17 (d, J=4.5 Hz, 1H), 3.97-3.91 (m, 1H), 2.31 (s, 3H), 1.31 (d, J=6.6 Hz, 3H).

Example 77

MS (ESI): m/z 178 [M+H]. 1H NMR (300 MHz, DMSO-d6) δ 8.5 (brs, 3H), 7.41 (s, 1H), 7.19 (d, J=7.8 Hz, 1H), 7.07 (d, J=8.1 Hz, 1H), 4.71 (s, 2H), 4.11 (d, J=3.6 Hz, 2H), 2.31 (s, 3H), 1.27 (d, J=6.3 Hz, 3H).

TABLE 8 Compounds prepared as described in Example 76 using the appropriately substituted starting materials. Example Structure Characterization Data 78 MS (ESI): m/z 192 [M + H]. 1H NMR (300 M Hz, DMSO-d6) δ 8.17 (brs, 3H), 7.18 (d, J = 7.8 Hz, 1H), 6.88 (d, J = 7.8 Hz, 1H), 4.86-4.70 (m, 2H), 4.36 (s, 1H), 3.90-3.83 (m, 1H), 2.23 (s, 3H), 2.21 (s, 3H), 1.32 (d, J = 6.6 Hz, 3H). 79 MS (ESI): m/z 192 [M + H]. 1H NMR (300 M Hz, DMSO-d6) δ 8.36 (brs, 3H), 7.19 (d, J = 7.8 Hz, 1H), 6.90 (d, J = 7.8 Hz, 1H), 4.74-4.20 (d, J = 5.1 Hz, 2H), 4.43-4.33 (m, 2H), 2.25 (s, 6H), 1.11 (d, J = 6.6 Hz, 3H). 80 MS (ESI): m/z 198 [M + H]. 1H NMR (300 M Hz, CD3OD) δ 7.47-7.31 (m, 2H), 7.28-7.17 (m, 1H), 5.01-4.78 (m, 2 H), 4.45 (d, J = 2.1 Hz, 1H), 4.05- 3.89 (m, 1H), 1.41 (d, J = 6 Hz, 3H). 81 MS (ESI): m/z 198 [M + H]. 1H NMR (300 M Hz, DMSO-d6) δ 8.55 (brs, 3H), 7.50-7.42 (m, 2H), 7.22 (dd, J = 3 Hz, 6 Hz, 1H), 4.85-4.71 (m, 2H), 4.51- 4.44 (m, 1H), 4.32 (d, J = 3 Hz, 1H), 1.15 (d, J = 6 Hz, 3H). 82 MS (ESI): m/z 178 [M + H]. 1H NMR (300 M Hz, DMSO-d6) δ 8.13 (brs, 3H), 7.29 (t, J = 7.5 Hz, 1H), 7.16 (d, J = 7.5 Hz, 1H), 6.99 (d, J = 7.5 Hz, 1H), 4.92-4.76 (m, 2H), 4.34-4.32 (m, 1H), 3.95- 3.89 (m, 1H), 2.37 (s, 3H), 1.33 (d, J = 6.6 Hz, 3H). 83 MS (ESI): m/z 178 [M + H]. 1H NMR (300 M Hz, DMSO-d6) δ 8.42 (s, 1H), 7.30 (d, J = 9 Hz, 1H), 7.15 (d, J = 9 Hz, 1H), 7.00 (d, J = 9 Hz, 1H), 4.83- 4.69 (m, 2H), 4.46-4.39 (m, 1H), 4.26 (s, 1H), 2.41 (s, 3H), 1.12 (d, J = 9 Hz, 3H). 84 MS (ESI): m/z 194 [M + H]. 1H NMR (300 M Hz, CD3OD) δ 7.09 (d, J = 8.4 Hz, 1H), 7.01-6.97 (m, 1H), 4.92-4.82 (m, 2H), 4.19 (s, 1H), 4.02-3.99 (m, 1H), 3.82 (s, 3H), 1.38 (d, J = 6.9 Hz, 3H). 85 MS (ESI): m/z 194 [M + H]. 1H NMR (300 M Hz, CD3OD) δ 7.12-7.08 (m, 1H), 7.02-6.97 (m, 2H), 4.78 (s, 2H), 4.25-4.20 (m, 1H), 4.14 (d, J = 2.7 Hz, 1H), 3.82 (s,3H), 1.30 (d, J = 6.6 Hz, 3H). 86 MS (ESI): m/z 198 [M + H]. 1H NMR (300 M Hz, CD3OD) δ 7.50-7.47 (m, 1H), 7.41-7.37 (m, 1H), 5.02 (d, J = 16.5 Hz, 1H), 4.77 (d, J = 16.2 Hz, 1H), 4.29 (s, 1H), 4.04-3.99 (m, 1H), 1.41 (d, J = 6.6 Hz, 3H). 87 MS (ESI): m/z 198 [M + H]. 1H NMR (300 M Hz, CD3OD) δ 7.52-7.48 (m, 1H), 7.43-7.39 (m, 2H), 4.90-4.73 (m, 2H)-4.33-4.29 (m, 1H), 4.23 (d, J = 2.1 Hz, 1H), 1.29 (d, J = 6.9 Hz, 3H). 88 MS (ESI): m/z 182 [M + H]. 1H NMR (300 M Hz, DMSO-d6) δ 8.40 (brs, 3H), 7.43 (dd, J = 8.4 Hz, 1.5 Hz, 1H), 7.29-7.25 (m, 2H), 4.87 (d, J = 12 Hz, 1H), 4.76 (d, J = 11.7 Hz, 1H), 4.27 (s, 1H), 4.00- 3.94 (m, 1H), 1.33 (d, J = 4.8 Hz, 3H). 89 MS (ESI): m/z 182 [M + H]. 1H NMR (300 M Hz, DMSO-d6) δ 8.64 (brs, 3H), 7.53 (d, J = 8.0 Hz, 1H), 7.25 (dd, J = 1.8 Hz, 8.0 Hz, 2H), 4.81-4.71 (m, 2H), 4.21 (d, J = 3.0 Hz, 1H), 4.11-4.08 (m, 1H), 1.31 (t, J = 4.8 Hz, 3H). 90 MS (ESI): m/z 214 [M + H]. 1H NMR (300 M Hz, CD3OD) δ 8.13 (d, J = 8.0 Hz, 1H), 8.00 (d, J = 8.0 Hz, 2H), 7.72-7.70 (m, 1H), 7.64-7.62 (m, 1H), 7.31 (d, J = 8.0 Hz, 1H), 5.04 (s, 2H), 4.97 (s, 1H), 4.59-4.57 (m, 1H), 1.35 (d, J = 4.0 Hz, 3H) . 91 MS (ESI): m/z 214 [M + H]. 1H NMR (300 M Hz, CD3OD) δ 8.10 (d, J = 8.0 Hz, 1H), 7.99 (d, J = 8.0 Hz, 2H), 7.72-7.70 (m, 1H), 7.64-7.62 (m, 1H), 7.31 (d, J = 8.0 Hz, 1H), 5.13-5.10 (m, 2H), 5.02 (s, 1H), 4.20-4.18 (m, 1H), 1.55 (d, J = 4.0 Hz, 3H). 92 MS (ESI): m/z 232 [M + H]. 1H NMR (300 M Hz, CD3OD) δ 7.64 (d, J = 6.3 Hz, 1H), 7.20 (d, J = 6.3 Hz, 1H), 4.84 (s, 2H), 4.51 (s, 1H), 4.45-4.40 (m, 1H), 1.27 (d, J = 6.3 Hz, 3H). 93 MS (ESI): m/z 198 [M + H]. 1H NMR (300 M Hz, CD3OD) δ 7.43 (d, J = 8.4 Hz, 1H), 7.37-7.33 (m, 1H), 7.25-7.19 (m, 1H), 4.97-4.80 (m, 2H), 4.28- 4.25 (m, 1H), 4.05-3.98 (m, 1H), 1.39 (d, J = 6.6 Hz, 3H). 94 MS (ESI): m/z 198 [M + H]. 1H NMR (300 M Hz, CD3OD) δ 7.44-7.35 (m, 2H), 7.26 (s, 1H), 4.82 (s, 2H), 4.31-4.23(m, 1H), 4.24-4.18 (m, 1H), 1.31 (d, J = 6.6 Hz, 3H). 95 MS (ESI): m/z 178 [M + H]. 1H NMR (300 M Hz, CD3OD) δ 7.29 (d, J = 7.8 Hz, 1H), 7.16 (d, J = 7.8 Hz, 1H), 7.00 (s, 1H), 4.92 (d, J = 15.6 Hz, 1H), 4.82 (d, J = 15.6 Hz, 1H), 4.20 (s, 1H), 4.04-3.91 (m, 1H), 2.34 (s, 3H), 1.38 (d, J = 6.6 Hz, 3H). 96 MS (ESI): m/z 178 [M + H]. 1H NMR (300 M Hz, CD3OD) δ 7.31 (d, J = 7.8 Hz, 1H), 7.16 (d, J = 7.8 Hz, 1H), 7.00 (s, 1H), 4.80 (s, 2H), 4.28-4.21 (m, 1H), 4.13 (s, 1H), 2.34 (s, 3H), 1.29 (d, J = 6.9 Hz, 3H). 97 MS (ESI): m/z 178 [M + H]. 1H NMR (300 M Hz, DMSO-d6) δ 8.23 (brs, 3H), 7.31-7.20 (m, 3H), 4.82 (d, J = 15.9 Hz, 1H), 4.69 (d, J = 15.9 Hz, 2H), 4.22-4.21 (m, 1H), 3.97-3.90 (m, 1H), 2.14 (s, 3H), 1.31 (d, J = 6.6 Hz, 3H). 98 MS (ESI): m/z 178 [M + H]. 1H NMR (300 M Hz, DMSO-d6) δ 8.54 (brs, 3H), 7.42 (d, J = 8.4 Hz, 1H), 7.27-7.18 (m, 2H), 4.68 (s, 2H), 4.20-4.12 (m, 2H), 2.15 (s, 3H), 1.24 (d, J = 6.3 Hz, 3H). 194 MS (ESI): m/z 198 [M + H]. 1H NMR (300 M Hz, CD3OD) δ 7.50 (d, J = 2.1 Hz, 1H), 7.43 (dd, J = 8.3, 2.1 Hz, 1H), 7.20 (d, J = 8.3 Hz, 1H), 4.95 (d, J = 15.8 Hz, 1H), 4.83 (d, J = 15.8 Hz, 1H), 4.28 (s, 1H), 4.03 (m, 1H), 1.40 (d, J = 6.6 Hz, 3H). 196 MS (ESI): m/z 218 [M + H]. 1H NMR (300 M Hz, CD3OD) δ 7.11 (d, J = 7.9 Hz, 1H), 6.89 (d, J = 7.8 Hz, 1H), 4.90 (d, J = 15.5 Hz, 1H), 4.83 (d, J = 15.5 Hz, 1H), 4.34 (s, 1H), 3.96 (m, 1H), 2.86-2.73 (m, 4H), 1.92-1.72 (m, 4H), 1.39 (d, J = 6.6 Hz, 3H). 197 MS (ESI): m/z 218 [M + H]. 1H NMR (300 M Hz, CD3OD) δ 7.11 (d, J = 7.9 Hz, 1H), 6.89 (d, J = 7.8 Hz, 1H), 4.90 (d, J = 15.5 Hz, 1H), 4.83 (d, J = 15.5 Hz, 1H), 4.34 (s, 1H), 3.96 (m, 1H), 2.86-2.73 (m, 4H), 1.92-1.72 (m, 4H), 1.39 (d, J = 6.6 Hz, 3H). 212 MS (ESI): m/z 182 [M + H]. 1H NMR (300 M Hz, D2O) δ 7.42 (dd, J = 8.6, 5.5 Hz, 1H), 7.08 (td, J = 8.7, 2.6 Hz, 1H), 6.94 (dd, J = 9.4, 2.5 Hz, 1H), 4.99-4.86 (m, 2H), 4.35 (s, 1H), 4.16-4.08 (m, 1H), 1.37 (d, J = 6.7 Hz, 3H). 213 MS (ESI): m/z 182 [M + H]. 1H NMR (300 M Hz, D2O) δ 7.42 (dd, J = 8.6, 5.5 Hz, 1H), 7.10 (td, J = 8.7, 2.7 Hz, 1H), 6.95 (dd, J = 9.3, 2.6 Hz, 1H), 4.89-4.82 (m, 2H), 4.42-4.35 (m, 1H), 4.29 (d, J = 1.7 Hz, 1H), 1.26 (d, J = 6.7 Hz, 3H). 214 MS (ESI): m/z 182 [M + H]. 1H NMR (300 M Hz, CD3OD) δ 7.45-7.38 (m, 1H), 7.30 (d, J = 7.5 Hz, 1H), 7.24-7.11 (m, 1H), 4.88-4.78 (m, 2H), 4.35- 4.21 (m, 2H), 1.32 (d, J = 6.7 Hz, 3H). 215 MS (ESI): m/z 182 [M + H]. 1H NMR (300 M Hz, CD3OD) δ 7.45-7.35 (m, 1H), 7.27 (d, J = 7.5 Hz, 1H), 7.22-7.12 (m, 1H), 5.05 (d, J = 16.1 Hz, 1H), 4.83 (d, J = 16.1 Hz, 1H), 4.31 (s, 1H), 4.07-4.00 (m, 1H), 1.41 (d, J = 6.6 Hz, 3H). 216 MS (ESI): m/z 182 [M + H]. 1H NMR (300 M Hz, CD3OD) δ 7.41-7.43 (m, 1H), 7.14 (t, J = 9.0 Hz, 1H), 7.06 (d, J = 7.7 Hz, 1H), 4.86 (s, 2H), 4.45- 4.27 (m, 2H), 1.30 (d, J = 6.7 Hz, 3H). 217 MS (ESI): m/z 182 [M + H]. 1H NMR (300 M Hz, CD3OD) δ 7.50-7.42 (m, 1H), 7.12 (t, J = 9.0 Hz, 1H), 7.04 (d, J = 7.8 Hz, 1H), 4.99 (d, J = 15.9 Hz, 1H), 4.86 (d, J = 16.0 Hz, 1H), 4.49 (s, 1H), 4.04- 3.97 (m, 1H), 1.42 (d, J = 6.6 Hz, 3H). 219 MS (ESI): m/z 192 [M + H]. 1H NMR (300 M Hz, CD3OD) δ 7.28-7.25 (m, 2H), 7.09 (d, J = 8.5 Hz, 1H), 4.94 (d, J = 12.3 Hz, 1H), 4.83 (d, J = 15.4 Hz, 1H), 4.21 (brs, 1H), 4.07-3.98 (m, 1H), 2.71- 2.63 (m, 2H), 1.39 (d, J = 6.6 Hz, 3H), 1.25 (t, J = 7.6 Hz, 3H). 220 MS (ESI): m/z 192 [M + H]. 1H NMR (300 M Hz, CD3OD) δ 7.28-7.25 (m, 2H), 7.10 (d, J = 8.4 Hz, 1H), 4.81 (s, 2H), 4.29-4.21 (m, 1H), 4.15 (d, J = 2.5 Hz, 1H), 2.71-2.63 (m, 2H), 1.31 (d, J = 6.7 Hz, 3H), 1.24 (t, J = 7.8 Hz, 3H). 225 MS (ESI): m/z 212 [M + H]. 1H NMR (300 M Hz, CD3OD) δ 7.35 (d, J = 8.2 Hz, 1H), 7.23 (d, J = 8.2 Hz, 1H), 4.99 (d, J = 12.1 Hz, 1H), 4.76 (d, J = 16.4 Hz, 1H), 4.38 (s, 1H), 4.00-3.94 (m, 1H), 2.39 (s, 3H), 1.41 (d, J = 6.6 Hz, 3H). 228 MS (ESI): m/z 232 [M + H]. 1H NMR (300 M Hz, CD3OD) δ 7.81 (s, 1H), 7.73 (d, J = 8.1 Hz, 1H), 7.42 (d, J = 8.1 Hz, 1H), 4.99 (d, J = 16.5 Hz, 1H), 4.90 (d, J = 16.5 Hz, 1H), 4.39 (brs, 1H), 4.11-4.03 (m, 1H), 1.42 (d, J = 6.6 Hz, 3H). 229 MS (ESI): m/z 232 [M + H]. 1H NMR (300 M Hz, CD3OD) δ 7.82 (s, 1H), 7.73 (d, J = 8.1 Hz, 1H), 7.42 (d, J = 8.1 Hz, 1H), 4.91 (s, 2H), 4.35-4.28 (m, 2H), 1.30 (d, J = 6.6 Hz, 3H). 230 MS (ESI): m/z 194 [M + H]. 1H NMR (300 M Hz, CD3OD) δ 7.39 (t, J = 8.1 Hz, 1H), 6.95 (d, J = 8.1 Hz, 1H), 6.77 (d, J = 7.8 Hz, 1H), 4.91 (d, J = 15.3 Hz, 1H), 4.81 (d, J = 15.6 Hz, 1H), 4.38 (brs, 1H), 4.03-3.93 (m, 1H), 3.93 (s, 3H), 1.40 (d, J = 6.7 Hz, 3H). 231 MS (ESI): m/z 194 [M + H]. 1H NMR (300 M Hz, CD3OD) δ 7.39 (t, J = 8.0 Hz, 1H), 6.97 (d, J = 8.3 Hz, 1H), 6.78 (d, J = 7.8 Hz, 1H), 4.86-4.70 (m, 2H), 4.38-4.19 (m, 2H), 3.92 (s, 3H), 1.30 (d, J = 6.7 Hz, 3H). 232 MS (ESI): m/z 194 [M + H]. 1H NMR (300 M Hz, D2O) δ 7.35 (d, J = 8.6 Hz, 1H), 6.95 (dd, J = 8.6, 2.6 Hz, 1H), 6.79 (d, J = 2.4 Hz, 1H), 4.83 (brs, 2H), 4.41-4.33 (m, 1H), 4.26 (s, 1H), 3.80 (s, 3H), 1.26 (d, J = 6.7 Hz, 3H). 233 MS (ESI): m/z 194 [M + H]. 1H NMR (300 M Hz, CD3OD) δ 7.33 (d, J = 8.6 Hz, 1H), 6.92 (dd, J = 8.6, 2.6 Hz, 1H), 6.76 (d, J = 2.5 Hz, 1H), 4.90-4.85 (m, 2H), 4.29 (s, 1H), 4.13-4.06 (m, 1H), 3.78 (s, 3H), 1.35 (d, J = 6.7 Hz, 3H). 234 MS (ESI): m/z 194 [M + H]. 1H NMR (300 M Hz, CD3OD) δ 7.34 (t, J = 8.0 Hz, 1H), 7.01 (dd, J = 8.0, 4.2 Hz, 2H), 4.95 (d, J = 16.2 Hz, 1H), 4.69 (d, J = 16.3 Hz, 1H), 4.22 (s, 1H), 4.02-3.95 (m, 1H), 3.84 (s, 3H), 1.39 (d, J = 6.6 Hz, 3H). 235 MS (ESI): m/z 194 [M + H]. 1H NMR (300 M Hz, CD3OD) δ 7.36 (t, J = 8.0 Hz, 1H), 7.03 (dd, J = 7.9, 3.4 Hz, 2H), 4.80 (d, J = 16.8 Hz, 1H), 4.70 (d, J = 16.5 Hz, 1H), 4.26-4.21 (m, 1H), 4.15 (s, 1H), 3.85 (s, 3H), 1.29 (d, J = 6.7 Hz, 3H). 237 MS (ESI): m/z 192 [M + H]. 1H NMR (300 M Hz, CD3OD) δ 7.36 (t, J = 8.0 Hz, 1H), 7.03 (dd, J = 7.9, 3.4 Hz, 2H), 4.80 (d, J = 16.8 Hz, 1H), 4.70 (d, J = 16.5 Hz, 1H), 4.26-4.21 (m, 1H), 4.15 (s, 1H), 3.85 (s, 3H), 1.29 (d, J = 6.7 Hz, 3H). 238 MS (ESI): m/z 192 [M + H]. 1H NMR (300 M Hz, DMSO-d6) δ 8.30 (s, 3H), 7.27 (s, 1H), 6.92 (s, 1H), 4.81-4.67 (m, 2H), 4.13-4.12 (m, 1H), 3.98- 3.83 (m, 1H), 2.21 (s, 6H), 1.31 (d, J = 6.6 Hz, 3H). 200 MS (ESI): m/z 212 [M + H]. 1H NMR (300 M Hz, CD3OD) δ 7.38 (d, J = 8.2 Hz, 1H), 7.26 (d, J = 8.0 Hz, 1H), 4.90 (d, J = 17.0 Hz, 1H), 4.72 (d, J = 17.0 Hz, 1H), 4.47-4.30 (m, 2H), 2.42 (s, 3H), 1.22 (d, J = 6.7 Hz, 3H). 248 MS (ESI): m/z 220 [M + H]. 1H NMR (300 M Hz, DMSO-d6) δ 8.15 (br.s., 3H), 6.89 (d, J = 8.4 Hz, 1H), 6.80 (d, J = 8.4 Hz, 1H), 4.82 (d, J = 15.3 Hz, 1H), 4.71 (d, J = 15.0 Hz, 1H), 4.23 (s, 1H), 4.11- 4.08 (m, 2H), 3.89-3.86 (m, 1H), 2.79-2.73 (m, 2H), 1.99-1.92 (m, 2H), 1.31 (d, J = 6.6 Hz, 3H). 249 MS (ESI): m/z 220 [M + H]. 1H NMR (300 M Hz, CD3OD) δ 6.89 (d, J = 8.4 Hz, 1H), 6.83 (d, J = 8.7 Hz, 1H), 4.78 (s, 2H), 4.38-4.2 (m, 1H), 4.27 (s, 1H), 4.16 (t, J = 5.4 Hz, 2H), 2.82 (t, J = 6.3 Hz, 2H), 2.12-2.00 (m, 2H), 1.24 (d, J = 6.9 Hz, 3H).

Example 99. trans-3-Ethyl-6-methylisochroman-4-amine hydrochloride

The compound of Example 99 was prepared as previously described in Example 76, using pent-1-en-3-ol. MS (ESI): m/z 192 [M+H]. 1H NMR (300 MHz, DMSO-d6) δ 8.39 (brs, 3H), 7.34 (s, 1H), 7.19 (d, J=7.8 Hz, 1H), 7.06 (d, J=7.8 Hz, 1H), 4.69 (s, 2H), 4.18 (s, 1H), 3.90-3.81 (m, 1H), 2.31 (s, 3H), 1.77-1.51 (m, 2H), 0.96 (t, J=5.4 Hz, 3H).

Example 100. cis-3-Ethyl-6-methylisochroman-4-amine hydrochloride

The compound of Example 100 was prepared as previously described in Example 76, using pent-1-en-3-ol. MS (ESI): m/z 192 [M+H]. 1H NMR (300 MHz, DMSO-d6) δ 8.18 (brs, 3H), 7.29 (s, 1H), 7.21 (d, J=7.8 Hz, 1H), 7.07 (d, J=7.8 Hz, 1H), 4.87 (d, J=11.7 Hz, 1H), 4.73 (d, J=11.7 Hz, 1H), 4.35 (s, 1H), 3.65-3.62 (m, 1H), 2.31 (s, 3H), 1.66-1.59 (m, 2H), 0.99 (t, J=5.4 Hz, 3H).

TABLE 9 Compounds prepared as described in Examples 99 and 100 using the appropriately substituted starting materials. Example Structure Characterization Data 198 MS (ESI): m/z 178 [M + H]. 1H NMR (300 MHz, CD3OD) δ 7.43-7.39 (m, 2H), 7.37-7.28 (m, 1H), 7.19 (d, J = 7.7 Hz, 1H), 5.00 (d, J = 15.6 Hz, 1H), 4.87 (d, J = 15.6 Hz, 1H), 4.31 (s, 1H), 3.79- 3.69 (m, 1H), 1.78-1.61 (m, 2H), 1.12 (t, J = 7.4 Hz, 3H). 199 MS (ESI): m/z 178 [M + H]. 1H NMR (300 MHz, CD3OD) δ 7.43-7.32 (m, 3H), 7.19 (d, J = 7.2 Hz, 1H), 4.82 (s, 2H), 4.22 (d, J = 2.2 Hz, 1H), 4.00-3.95 (m, 1H), 1.71-1.46 (m, 2H), 1.07 (t, J = 7.4 Hz, 3H). 201 MS (ESI): m/z 196 [M + H]. 1H NMR (300 MHz, CD3OD) δ 7.27-7.13 (m, 3H), 4.99 (d, J = 15.5 Hz, 1H), 4.82 (d, J = 15.4 Hz, 1H), 4.34 (s, 1H), 3.75-3.69(m, 1H), 1.79-1.58 (m, 2H), 1.11 (t, J = 7.4 Hz, 3H). 202 MS (ESI): m/z 196 [M + H]. 1H NMR (300 MHz, CD3OD) δ 7.30-7.11 (m, 3H), 4.77 (s, 2H), 4.25 (d, J = 2.3 Hz, 1H), 4.00-3.94 (m, 1H), 1.76-1.45 (m, 2H), 1.07 (t, J = 7.4 Hz, 3H). 207 MS (ESI): m/z 192 [M + H]. 1H NMR (300 MHz, CD3OD) δ 7.30 (t, J = 7.6 Hz, 1H), 7.19 (d, J = 7.3 Hz, 1H), 7.00 (d, J = 7.6 Hz, 1H), 4.88 (d, J = 16.2 Hz, 1H), 4.78 (d, J = 16.2 Hz, 1H), 4.37 (d, J = 1.3 Hz, 1H), 4.08-4.03 (m, 1H), 2.42 (s, 3H), 1.66-1.38 (m, 2H), 1.05 (t, J = 7.4 Hz, 3H). 208 MS (ESI): m/z 192 [M + H]. 1H NMR (300 MHz, CD3OD) δ 7.30 (t, J = 7.6 Hz, 1H), 7.20 (d, J = 7.2 Hz, 1H), 7.00 (d, J = 7.6 Hz, 1H), 5.01 (d, J = 15.7 Hz, 1H), 4.85 (d, J = 15.8 Hz, 1H), 4.42 (s, 1H), 3.72-3.67 (m, 1H), 2.42 (s, 3H), 1.81-1.59 (m, 2H), 1.12 (t, J = 7.4 Hz, 3H). 209 MS (ESI): m/z 212 [M + H]. 1H NMR (300 MHz, CD3OD) δ 7.49-7.39 (m, 2H), 7.19-7.16 (m, 1H), 4.83 (d, J = 6.5 Hz, 2H), 4.48 (brs, 1H), 4.13-4.07 (m, 1H), 1.67-1.39 (m, 2H), 1.06 (t, J = 7.4 Hz, 3H). 210 MS (ESI): m/z 206 [M + H]. 1H NMR (300 MHz, CD3OD) δ 7.25-7.22 (m, 2H), 7.06 (d, J = 8.1 Hz, 1), 4.79 (s, 2H), 4.37 (d, J = 1.9 Hz, 1H), 3.59 (dd, J = 9.7, 2.0 Hz, 1H), 2.36 (s, 3H), 1.82-1.72 (m, 1H), 1.09 (d, J = 6.5 Hz, 3H), 0.94 (d, J = 6.6 Hz, 3H). 211 MS (ESI): m/z 206 [M + H]. 1H NMR (300 MHz, CD3OD) δ 7.25-7.22 (m, 2H), 7.07 (d, J = 8.4 Hz, 1H), 4.98 (d, J = 15.3 Hz, 1H), 4.80 (d, J = 15.3 Hz, 1H), 4.43 (s, 1H), 3.36-3.31 (m, 1H), 2.36 (s, 3H), 1.86-1.76 (m, 1H), 1.15 (d, J = 6.4 Hz, 3H), 1.04 (d, J = 6.7 Hz, 3H). 218 MS (ESI): m/z 212 [M + H]. 1H NMR (300 MHz, CD3OD) δ 7.49-7.38 (m, 2H), 7.22-7.15 (m, 1H), 5.02 (d, J = 15.9 Hz, 1H), 4.85 (d, J = 16.2 Hz, 1H), 4.55 (brs, 1H), 3.76-3.68 (m, 1H), 1.79-1.66 (m, 2H), 1.13 (t, J = 7.4 Hz, 3H). 223 MS (ESI): m/z 206 [M + H]. 1H NMR (300 MHz, CD3OD) δ 7.46-7.30 (m, 3H), 7.19 (d, J = 7.2 Hz, 1H), 4.98 (d, J = 15.6 Hz, 1H), 4.86 (d, J = 15.6 Hz, 1H), 4.26 (brs, 1H), 3.95-3.90 (m, 1H), 1.97-1.89 (m, 1H), 1.70-1.60 (m, 1H), 1.48-1.39 (m, 1H), 1.03 (d, J = 6.6 Hz, 3H), 1.00 (d, J = 6.6 Hz, 3H). 224 MS (ESI): m/z 206 [M + H]. 1H NMR (300 MHz, CD3OD) δ 7.49-7.29 (m, 3H), 7.19 (d, J = 7.6 Hz, 1H), 4.82 (s, 2H), 4.30-4.13 (m, 2H), 1.94- 1.75 (m, 1H), 1.65-1.55 (m, 1H), 1.28-1.19 (m, 1H), 0.98 (t, J = 6.3 Hz, 6H). 250 MS (ESI): m/z 192 [M + H]. 1H NMR (300 MHz, DMSO-d6) δ 8.19 (brs, 3H), 7.47 (d, J = 7.5 Hz, 1H), 7.40-7.32 (m, 2H), 7.18 (d, J = 7.5 Hz, 1H), 4.90 (d, J = 15.6 Hz, 1H), 4.79 (d, J = 15.6 Hz, 1H), 4.28 (s, 1H), 3.78 (m, 1H), 1.60-1.56 (m, 2H), 1.52-1.44 (m, 2H), 0.95 (t, J = 7.2 Hz, 3H). 251 MS (ESI): m/z 192 [M + H]. 1H NMR (300 MHz, DMSO-d6) δ 8.42 (brs, 3H), 7.52 (d, J = 7.5 Hz, 1H), 7.38-7.33 (m, 2H), 7.18 (d, J = 7.5 Hz, 1H), 4.73 (m, 2H), 4.20 (s, 1H), 4.00 (m, 1H), 1.52- 1.47 (m, 4H), 0.91 (t, J = 6.9 Hz, 3H).

Example 101. 3,3-Dimethylisochroman-4-amine hydrochloride

The compound of Example 101 was prepared as previously described in Example 76, using 2-methylbut-3-en-2-ol. MS (ESI): m/z 178 [M+H]. 1H NMR (300 MHz, DMSO-d6) δ 8.35 (brs, 3H), 7.49 (d, J=6.9 Hz, 1H), 7.41-7.30 (m, 2H), 7.18 (d, J=7.2 Hz, 1H), 4.86-4.71 (m, 2H), 4.14 (s, 1H), 1.33 (s, 3H), 1.19 (s, 3H).

Example 104. cis-1-Methylisochroman-4-amine hydrochloride Example 105. trans-1-Methylisochroman-4-amine hydrochloride

The compounds are Example 104 and Example 105 were prepared as described previously in Example 76 using 1-(1-(allyloxy)ethyl)-2-bromobenzene (prepared as previously described in Example 21 using 1-(2-bromophenyl)ethanol).

Example 104. cis-1-Methylisochroman-4-amine hydrochloride

MS (ESI): m/z 164 [M+H]. 1H NMR (300 MHz, DMSO-d6) δ 8.66 (brs, 3H), 7.58 (d, J=5.4 Hz, 1H), 7.41-7.31 (m, 2H), 7.25 (d, J 5.7 Hz, 1H), 4.98 (q, J 5.1 Hz, 1H), 4.35 (t, J=2.7, 1H), 4.17 (dd, J=2.7 Hz, 8.7 Hz, 1H), 3.89 (dd, J=3 Hz, 9 Hz, 1H), 1.46 (d, J 5.1 Hz, 3H).

Example 105. trans-(1S,4S)-1-Methylisochroman-4-amine hydrochloride

MS (ESI): m/z 164 [M+H]. 1H NMR (300 MHz, DMSO-d6) δ 8.54 (brs, 3H), 7.51 (d, J=7.8 Hz, 1H), 7.41-7.28 (m, 3H), 4.78 (q, J=6.3 Hz, 1H), 4.31-4.29 (m, 1H), 4.21 (d, J=12.6 Hz, 1H), 3.87 (dd, J=2.1 Hz, 12.6 Hz, 1H), 1.53 (d, J=6.6 Hz, 3H).

TABLE 10 Compounds prepared as described in Examples 104 and 105 using the appropriately substituted starting materials. Example Structure Characterization Data 106 MS (ESI): m/z 178 [M + H]. 1H NMR (300 MHz, CDCl3) δ 8.67 (brs, 3H), 7.43 (s, 1H), 7.12 (d, J = 8.1 Hz, 1H), 6.95 (d, J = 7.8 Hz, 1H), 5.04-4.98 (m, 1H), 4.30 (s, 1H), 4.10 (s, 2H), 2.30 (s, 3H), 1.45 (d, J = 6.6 Hz, 3H). 107 MS (ESI): m/z 178 [M + H]. 1H NMR (300 MHz, CDCl3) δ 8.69 (brs, 3H), 7.46 (s, 1H), 7.14 (d, J = 8.1 Hz, 1H), 7.01 (d, J = 8.1 Hz, 1H), 4.80-4.74 (m, 1H), 4.50 (d, J = 12.3 Hz, 1H), 4.30 (s, 1H), 3.80 (dd, J = 1.8 Hz, 12.6 Hz, 1H), 2.30 (s, 3H), 1.59 (d, J = 6.6 Hz, 3H). 203 MS (ESI): m/z 198 [M + H]. 1H NMR (300 MHz, DMSO-d6) δ 8.60 (brs, 3H), 7.67 (d, J = 2.1 Hz, 1H), 7.46 (dd, J = 8.4, 2.2 Hz, 1H), 7.30 (d, J = 8.5 Hz, 1H), 4.97-4.93 (m, 1H), 4.39 (t, J = 3.7 Hz, 1H), 4.14 (dd, J = 12.4, 3.5 Hz, 1H), 3.85 (dd, J = 12.3, 4.2 Hz, 1H), 1.43 (d, J = 6.7 Hz, 3H). 204 MS (ESI): m/z 198 [M + H]. 1H NMR (300 MHz, DMSO-d6) δ 8.49 (brs, 3H), 7.61 (d, J = 2.2 Hz, 1H), 7.48 (dd, J = 8.4, 2.2 Hz, 1H), 7.34 (d, J = 8.5 Hz, 1H), 4.79-4.73 (m, 1H), 4.35 (s, 1H), 4.16 (d, J = 12.1 Hz, 1H), 3.86 (dd, J = 12.8, 2.3 Hz, 1H), 1.52 (d, J = 6.5 Hz, 3H). 205 MS (ESI): m/z 182 [M + H]. 1H NMR (300 MHz, D2O) δ 7.26-7.13 (m, 3H), 5.16-5.09 (m, 1H), 4.43 (s, 1H), 4.26 (dd, J = 13.1, 2.3 Hz, 1H), 3.98 (dd, J = 13.2, 2.1 Hz, 1H), 1.47 (d, J = 6.8 Hz, 3H). 206 MS (ESI): m/z 182 [M + H]. 1H NMR (300 MHz, D2O) δ 7.32-7.27 (m, 1H), 7.22-7.12 (m, 2H), 4.92-4.84 (m, 1H), 4.42 (s, 1H), 4.22 (d, J = 13.2 Hz, 1H), 4.02 (dd, J = 13.1, 2.0 Hz, 1H), 1.53 (d, J = 6.5 Hz, 3H). 221 MS (ESI): m/z 178 [M + H]. 1H NMR (300 MHz, D2O) δ 7.30 (t, J = 7.7 Hz, 1H), 7.17 (d, J = 7.4 Hz, 1H), 7.02 (d, J = 7.7 Hz, 1H), 5.18-5.10 (m, 1H), 4.54 (s, 1H), 4.18 (dd, J = 13.2, 1.6 Hz, 1H), 4.01 (dd, J = 13.2, 1.5 Hz, 1H), 2.33 (s, 3H), 1.44 (d, J = 6.8 Hz, 3H). 222 MS (ESI): m/z 178 [M + H]. 1H NMR (300 MHz, D2O) δ 7.32 (t, J = 7.7 Hz, 1H), 7.19 (d, J = 7.5 Hz, 1H), 7.10 (d, J = 7.8 Hz, 1H), 4.94-4.89 (m, 1H), 4.55 (s, 1H), 4.19 (d, J = 12.9 Hz, 1H), 3.93 (dd, J = 12.9, 1.3 Hz, 1H), 2.33 (s, 3H), 1.52 (d, J = 6.5 Hz, 3H). 226 MS (ESI): m/z 178 [M + H]. 1H NMR (300 MHz, D2O) δ 7.30 (t, J = 7.7 Hz, 1H), 7.18 (d, J = 7.6 Hz, 1H), 7.02 (d, J = 7.7 Hz, 1H), 5.18-5.11 (m, 1H), 4.54 (s, 1H), 4.18 (dd, J = 13.2, 1.6 Hz, 1H), 4.01 (dd, J = 13.2, 1.6 Hz, 1H), 2.33 (s, 3H), 1.44 (d, J = 6.8 Hz, 3H). 227 MS (ESI): m/z 178 [M + H]. 1H NMR (300 MHz, D2O) δ 7.32 (t, J = 7.7 Hz, 1H), 7.19 (d, J = 7.5 Hz, 1H), 7.11 (d, J = 7.7 Hz, 1H), 4.95-4.88 (m, 1H), 4.55 (s, 1H), 4.21 (dd, J = 12.9, 1.4 Hz, 1H), 3.94 (dd, J = 12.9, 1.7 Hz, 1H), 2.33 (s, 3H), 1.52 (d, J = 6.5 Hz, 3H).

Example 236. trans-N,1-dimethylisochroman-4-amine hydrochloride

The compound of Example 236 was prepared as previously described in Example 57. MS (ESI): m/z 178 [M+H]. 1H NMR (300 MHz, DMSO-d6) δ 9.26 (s, 1H), 9.11 (s, 1H), 7.54 (d, J=7.5 Hz, 1H), 7.45 (d, J=7.5 Hz, 1H), 7.38-7.31 (m, 2H), 4.82-4.80 (m, 1H), 4.40 (d, J=13.1 Hz, 1H), 4.27 (brs, 1H), 3.86 (dd, J=13.1, 2.0 Hz, 1H), 2.56 (t, J=5.4 Hz, 3H), 1.54 (d, J=6.5 Hz, 3H).

Example 239. cis-N,N,1-trimethylisochroman-4-amine hydrochloride

The compound of Example 239 was prepared as previously described in Example 60. MS (ESI): m/z 192 [M+H]. 1H NMR (300 MHz, CD3OD) δ 7.59-7.47 (m, 2H), 7.44-7.37 (m, 2H), 4.90-4.84 (m, 1H), 4.64 (d, J=14.1 Hz, 1H), 4.40 (s, 1H), 3.97 (dd, J=14.1, 2.4 Hz, 1H), 2.95 (s, 3H), 2.90 (s, 3H), 1.61 (d, J=6.5 Hz, 3H).

Example 240. cis-N,1-dimethylisochroman-4-amine hydrochloride

The compound of Example 240 was prepared as previously described in Example 57. MS (ESI): m/z 178 [M+H]. 1H NMR (300 MHz, CD3OD) δ 7.51-7.41 (m, 2H), 7.38-7.33 (m, 1H), 7.26 (d, J=7.7 Hz, 1H), 5.18-5.11 (m, 1H), 4.23-4.11 (m, 3H), 2.74 (s, 3H), 1.52 (d, J=6.8 Hz, 3H).

Example 256. cis-3-Isopropylisochroman-4-amine hydrochloride Example 257. trans-3-Isopropylisochroman-4-amine hydrochloride

Step 1.

A solution of 2-bromo-3-methylbutanoic acid (5 g, 27.6 mmol) in 100 mL of CH2Cl2 was chilled in an ice bath and treated with 1,1-carbonyldiimidazole (4.68 g, 28.9 mmol). The ice bath was removed and the reaction mixture was stirred at ambient temperature for 15 min. N, O-dimethylhydroxylamine (1.76 g, 28.9 mmol) was added to the reaction mixture and stirring at ambient temperature was maintained for 2 h. Water (100 mL) was added to the reaction vessel and the resulting biphasic mixture was transferred to a separatory funnel. The organic phase was washed with 10% aqueous citric acid (2×100 mL), saturated aqueous NaHCO3 (100 mL), and saturated aqueous NaCl (2×100 mL). The organic phase was dried over anhydrous Na2SO4, filtered, and concentrated under reduced pressure to provide 2-bromo-N-methoxy-N,3-dimethylbutanamide 256.1 (2.80 g, crude) as a yellow oil. GC-MS: m/z 223, 225 [M+H]. 1HNMR (300 MHz, CDCl3) δ 4.51 (d, J=9.3 Hz, 1H), 3.78 (s, 3H), 3.24 (s, 3H), 2.36-2.28 (m, 1H), 1.16 (d, J=6.9 Hz, 3H), 0.98 (d, J=6.9 Hz, 3H).

Step 2.

A solution of (2-bromophenyl)methanol (3.47 g, 18.6 mmol) in DMF (100 mL) was cooled in an ice bath and treated with sodium hydride (743 mg, 18.6 mmol, 95% in mineral oil). The resulting slurry was stirred for 30 min and then treated with Compound 256.1 (2.8 g, 12.4 mmol). The reaction was stirred at ambient temperature for 1 h and then partitioned between H2O (100 mL) and EtOAc (100 mL). The aqueous phase was extracted with EtOAc (2×100 mL). The combined organics were washed with saturated aqueous NaCl (2×100 mL), dried over anhydrous Na2SO4, filtered, and concentrated under reduced pressure. The resulting oil was purified by flash column chromatography (SiO2, 100% hexanes gradient to 10% EtOAc/hexanes (90%) to provide 2-((2-bromobenzyl)oxy)-N-methoxy-N,3-dimethylbutanamide 256.2 (4.05 g, 12.2 mmol) as a colorless oil. MS (ESI): m/z 330 [M+H],

Step 3.

A solution of Compound 256.2 (4.5 g, 13.6 mmol) in THF (100 mL) was chilled in a dry ice/acetone bath and treated with tert-butyllithium (33.9 mL, 1.7M in pentane). The reaction was stirred in the dry ice/acetone bath for 10 min. Excess base was quenched by the dropwise addition of saturated aqueous NH4Cl. The mixture was partitioned between saturated aqueous NH4Cl (100 mL) and EtOAc (100 mL). The aqueous phase was extracted with EtOAc (2×100 mL). The combined organics were dried over anhydrous Na2SO4, filtered, and concentrated under reduced pressure. The resulting oil was purified by flash column chromatography (SiO2, 100% hexanes) to afford 3-isopropylisochroman-4-one 256.3 (600 mg, 3.15 mmol) as a yellow oil. 1H NMR (300 MHz, CDCl3) δ 8.03 (d, J=7.8 Hz, 1H), 7.57-7.52 (m, 1H), 7.40 (t, J=7.5 Hz, 1H), 7.21 (d, J=7.8 Hz, 1H), 4.95 (d, J=15.0 Hz, 1H), 4.86 (d, J=15.0 Hz, 1H), 3.97-3.94 (m, 1H), 2.60-2.53 (m, 1H), 1.15 (d, J=7.5 Hz, 3H), 0.98 (d, J=7.5 Hz, 3H).

Step 4.

3-Isopropylisochroman-4-ol 256.4 was prepared as previously described in Example 1. GC-MS: m/z 192 [M+H]. 1H NMR (300 MHz, CDCl3) δ 7.54-7.52 (m, 0.4H), 7.43-7.30 (m, 0.6H), 7.31-7.23 (m, 2H), 7.04-6.99 (m, 1H), 4.90-4.49 (m, 3H), 3.33 (dd, J=7.5, 4.5 Hz, 0.4H), 3.08 (dd, J=9.6, 1.2 Hz, 0.6H), 2.13-2.07 (m, 1H), 1.94 (d, J=10.5 Hz, 0.6H), 1.81 (d, J=9.0 Hz, 0.4H), 1.22-1.02 (m, 6H).

Step 5.

4-Azido-3,isopropylisochroman 256.5 was prepared as previously described in Example 1. 1H NMR (300 MHz, CDCl3) δ 7.46-7.43 (m, 1H), 7.34-7.27 (m, 2H), 7.06-7.03 (m, 1H), 4.83-4.72 (m, 2H), 4.33 (d, J=7.5 Hz, 1H), 3.50-3.46 (m, 1H), 2.11-2.04 (m, 1H), 1.08-1.02 (m, 6H).

Step 6.

Compound s 256.5a and 256.5b were prepared as previously described in Example 73.

tert-butyl-(cis-3-isopropylisochroman-4-yl)carbamate 256.5a TLC higher Rf. 1H NMR (300 MHz, CDCl3) δ 7.49-7.46 (m, 1H), 7.25-7.22 (m, 2H), 7.00-6.97 (m, 1H), 5.02-4.72 (m, 4H), 3.17 (dd, J=9.6, 1.8 Hz, 1H), 1.91-1.83 (m, 1H), 1.42 (s, 9H), 1.08 (d, J=6.6 Hz, 3H), 0.99 (d, J=6.6 Hz, 3H).

tert-butyl-(trans-3-isopropylisochroman-4-yl)carbamate 256.5b TLC lower Rf. 1H NMR (300 MHz, CDCl3) δ 7.39-7.36 (m, 1H), 7.24-7.20 (m, 2H), 6.99-6.96 (m, 1H), 4.86-4.69 (m, 4H), 3.32 (dd, J=6.9, 5.1 Hz, 1H), 1.94-1.92 (m, 1H), 1.50 (s, 9H), 1.05 (d, J=6.9 Hz, 6H).

Step 7.

The compounds of Example 256 and Example 257 were prepared as previously described in Example 1.

Example 256

MS (ESI): m/z 292 [M+H]. 1H NMR (300 MHz, CD3OD) δ 7.49-7.36 (m, 3H), 7.23 (d, J=7.8 Hz, 1H), 5.07 (d, J=15.6 Hz, 1H), 4.94 (d, J=15.6 Hz, 1H), 4.53 (s, 1H), 3.40 (dd, J=10.2, 1.2 Hz, 1H), 1.92-1.85 (m, 1H), 1.20 (d, J=6.3 Hz, 3H), 1.09 (d, J=6.6 Hz, 3H).

Example 257

MS (ESI): m/z 292 [M+H]. 1H NMR (300 MHz, CD3OD) δ 7.49-7.38 (m, 3H), 7.23 (d, J=7.2 Hz, 1H), 4.88 (s, 2H), 4.47 (d, J=2.1 Hz, 1H), 3.66 (dd, J=9.9, 2.1 Hz, 1H), 1.87-1.80 (m, 1H), 1.14 (d, J=6.6 Hz, 3H), 0.98 (d, J=6.6 Hz, 3H).

Example 273. cis-3-Isopropylisochroman-4-amine hydrochloride Example 274. trans-3-Isopropylisochroman-4-amine hydrochloride

Step 1.

A solution of phenylacetylene (4 g, 39.1 mmol) in 50 mL of anhydrous THF was cooled in a dry ice/acetone bath under a N2 atmosphere. Butyllithium (18.7 mL, 46.9 mmol) was added in a dropwise fashion, and the reaction was stirred at −78° C. for 30 min. A solution of methyl 2,2-difluoroacetate (5.16 g, 46.9 mmol) in THF (5 mL) was added, followed by boron trifluoride etherate (5.91 mL, 46.9 mmol). The cold bath was removed, and the reaction was stirred at ambient temperature for 12 h. Saturated aqueous NH4Cl (50 mL) was added to the reaction vessel and the reaction mixture was extracted with EtOAc (3×50 mL). The combined organics were washed with saturated aqueous NaCl (2×80 mL), dried over anhydrous Na2SO4, filtered, and concentrated under reduced pressure. Purification by flash column chromatography (SiO2, 100% hexane) afford 1,1-difluoro-4-phenylbut-3-yn-2-one 273.1 (5.50 g, 30.5 mmol) as a yellow oil. GC-MS: m/z 180 [M+H]. 1H NMR (300 MHz, CDCl3) δ 7.67 (d, J=7.2 Hz, 1H), 7.57-7.52 (m, 1H), 7.46-7.41 (m, 2H), 5.88 (t, J=54.0 Hz, 1H).

Step 2.

Compound 273.1 (5.5 g, 30.5 mmol) was dissolved in THF (100 mL) and cooled in an ice bath. LiAl4 (1.73 g, 45.7 mmol) was added slowly in portions and the reaction mixture was stirred 0° C. for 30 min. Excess hydride was quenched by the dropwise addition of H2O (1.8 mL), followed by 1.8 mL of 15% aqueous NaOH. After stirring at room temperature for 0.5 hour, the solid was removed by filtration. The filtrate was concentrated to provide (E)-1,1-difluoro-4-phenylbut-3-en-2-ol 273.2 (5.30 g, 28.7 mmol) as a colorless oil. GC-MS: m/z 184 [M+H]. 1H NMR (300 MHz, CDCl3) δ 7.44-7.27 (m, 5H), 6.82 (d, J=15.9 Hz, 1H), 6.25-6.18 (dd, J=15.9, 6.3 Hz, 1H), 5.72 (td, J=56.1, 4.2 Hz, 1H), 4.53-4.43 (m, 1H), 2.25 (d, J=5.1 Hz, 1H).

Step 3.

Compound 273.3 was prepared as previously described in Example 10. 1HNMR (300 MHz, CDCl3) δ 7.58-7.50 (m, 2H), 7.49-7.43 (m, 1H), 7.39-7.25 (m, 4H), 7.21-7.15 (m, 2H), 6.81 (d, J=15.9 Hz, 1H), 6.18 (dd, J=16.2, 7.5 Hz, 1H), 5.84-5.64 (m, 1H), 4.79-4.63 (m, 2H), 4.23-4.19 (m, 1H).

Step 4.

Compound 273.4 was prepared as previously described in Example 10. 1H NMR (300 MHz, CDCl3) δ 7.66-7.62 (m, 1H), 7.44-7.38 (m, 3H), 7.35-7.29 (m, 3H), 7.25-7.16 (m, 1H), 7.08-7.05 (m, 1H), 6.06-5.67 (td, J=54.9, 5.4 Hz, 1H), 5.20-5.11 (m, 1H), 5.01 (d, J=15.3 Hz, 1H), 4.83 (d, J=15.3 Hz, 1H).

Step 5.

Compound 273.5 was prepared as previously described in Example 10. 1H NMR (300 MHz, CDCl3) δ 8.06 (d, J=6.9 Hz, 1H), 7.65-7.59 (m, 1H), 7.48-7.43 (m, 1H), 7.29-7.24 (m, 1H), 6.37 (td, J=53.4, 1.8 Hz, 1H), 5.14 (d, J=15.3 Hz, 1H), 5.02 (d, J=15.3 Hz, 1H), 4.51-4.41 (m, 1H).

Step 6.

Compound 273.6 was prepared as previously described in Example 1. 1H NMR (300 MHz, CDCl3) δ 7.58 (d, J=7.2 Hz, 0.5H), 7.43 (d, J=6.9 Hz, 0.5H), 7.35-7.30 (m, 2H), 7.09-7.02 (m, 1H), 6.24-5.86 (m, 1H), 4.98-4.65 (m, 3H), 3.81-3.74 (m, 1H), 2.34 (d, J=6.6 Hz, 0.5H), 2.12 (d, J=9.9 Hz, 0.5H).

Step 7.

Compound 273.7 was prepared as previously described in Example 1. 1H NMR (300 MHz, CDCl3) δ 7.51-7.48 (m, 0.5H), 7.43-7.32 (m, 2.5H), 7.15-7.06 (m, 1H), 6.22-5.82 (m, 1H), 5.04 (d, J=15.3 Hz, 0.5H), 4.91-4.86 (m, 1.5H), 4.62 (d, J=7.8 Hz, 0.5H), 4.34 (s, 0.5H), 3.96-3.88 (m, 1H).

Step 8.

Compound s 273.8a and 273.8b were prepared as previously described in Example 1.

tert-butyl-(cis-3-(difluoromethyl)isochroman-4-yl)carbamate 273.8a TLC higher Rf. 1H NMR (300 MHz, CDCl3) □ 7.44-7.41 (m, 1H), 7.34-7.22 (m, 2H), 7.05-7.00 (m, 1H), 6.11-5.73 (m, 1H), 4.99-4.95 (m, 2H), 4.88-4.81 (m, 2H), 3.91-3.85 (m, 1H), 1.44 (s, 9H).

tert-butyl-(trans-3-(difluoromethyl)isochroman-4-yl)carbamate 273.8b TLC lower Rf. 1H NMR (300 MHz, CDCl3) □ 7.42-7.40 (m, 1H), 7.32-7.26 (m, 2H), 7.04-7.01 (m, 1H), 6.16-5.78 (m, 1H), 5.02-4.95 (m, 1H), 4.90-4.89 (m, 2H), 4.85-4.80 (m, 1H), 3.84-3.80 (m, 1H), 1.44 (s, 9H).

Step 7.

The compounds of Example 273 and Example 274 were prepared as previously described in Example 1.

Example 273

MS (ESI): m/z 200 [M+H]. 1H NMR (300 MHz, CD3OD) δ 7.51-7.45 (m, 2H), 7.42-7.38 (m, 1H), 7.25 (d, J=7.5 Hz, 1H), 6.19 (td, J=54.0, 3.9 Hz, 1H), 5.15 (d, J=15.6 Hz, 1H), 5.02 (d, J=15.6 Hz, 1H), 4.67 (s, 1H), 4.34-4.24 (m, 1H).

Example 274

MS (ESI): m/z 200 [M+H]. 1H NMR (300 MHz, CD3OD) δ 7.54-7.40 (m, 3H), 7.25 (d, J=7.5 Hz, 1H), 6.13 (td, J=54.3, 5.1 Hz, 1H), 5.00 (s, 2H), 4.64 (d, J=2.4 Hz, 1H), 4.37-4.27 (m, 1H).

Example 275. cis-3-(fluoromethyl)isochroman-4-amine hydrochloride

The compound of Example 275 was prepared as previously described in Example 273 using ethyl 2-fluoroacetate. MS (ESI): m/z 182 [M+H]. 1H NMR (300 MHz, DMSO-d6) δ 8.41 (s, 3H), 7.52-7.50 (d, J=7.5 Hz, 1H), 7.44-7.31 (m, 2H), 7.21-7.19 (d, J=7.2 Hz, 1H), 5.0-4.85 (m, 2.5H), 4.83-4.68 (m, 1H), 4.62-4.56 (m, 0.5H), 4.45 (s, 1H), 4.21-4.15 (d, J=17.1 Hz, 1H).

Example 276. cis-3-(fluoromethyl)isochroman-4-amine hydrochloride

The compound of Example 276 was prepared as previously described in Example 274 using ethyl 2-fluoroacetate. MS (ESI): m/z 182 [M+H]. 1H NMR (300 MHz, DMSO-d6) δ 8.8 (s, 3H), 7.68-7.66 (m, 1H), 7.38-7.34 (m, 2H), 7.21-7.20 (m, 1H), 4.91-4.61 (m, 4H), 4.44 (s, 1H), 4.29-4.17 (m, 1H).

Example 277. 4-Aminoisochroman-6-yl ethyl(methyl)carbamate hydrochloride

Step 1.

6-Methoxyisochroman-4-one 10.3 (1 g, 5.61 mmol) was dissolved in anhydrous DMF (3 mL) under a N2 atmosphere and treated with sodium ethanethiolate (841 mg, 8.41 mmol). The reaction mixture was heated to 140° C. and stirred at that temperature for 15 min. After cooling to room temperature, the mixture was partitioned between H2O (15 mL) and EtOAc (10 mL). The aqueous phase was extracted with EtOAc (2×10 mL). The aqueous phase was acidified with aqueous 1N HCl until pH=2 and extracted with EtOAc (2×10 mL). The combined organics were dried over anhydrous Na2SO4, filtered, and concentrated under reduced pressure to afford 6-hydroxyisochroman-4-one 277.1 (696 mg, 4.24 mmol) as brown oil which was used without further purification. 1H NMR (300 MHz, CDCl3) δ 7.45 (d, I=2.1 Hz, 1H), 7.15-7.07 (m, 2H), 4.84 (s, 2H), 4.35 (s, 2H).

Step 2.

A solution of Compound 277.1 (696.5 mg, 4.23 mmol) in CH3CN (15 mL) was treated with ethyl(methyl)carbamic chloride (1.02 g, 8.46 mmol) and K2CO3 (876 mg, 6.34 mmol). The reaction mixture was heated to 55° C. and stirred at that temperature for 5 h. After concentration under reduced pressure, the residue was diluted with water (10 mL) and extracted with EtOAc (3×10 mL). The combined organics were dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure. Purification by flash column chromatography (SiO2, gradient elution from 10% EtOAc/hexanes to 75% EtOAc/hexanes) provided 4-oxoisochroman-6-yl ethyl(methyl)carbamate 277.2 (230 mg, 0.922 mmol) as a yellow oil. 1H NMR (300 MHz, CDCl3) δ 7.76 (s, 1H), 7.37 (d, J=8.1 Hz, 1H), 7.23 (d, J=8.4 Hz, 1H), 4.89 (s, 2H), 4.36 (s, 2H), 3.50-3.40 (m, 2H), 3.04 (d, J=24.3 Hz, 3H), 1.28-1.11 (m, 3H).

Step 3.

Compound 277.2 (230 mg, 922 μmol) was dissolved in MeOH (3 mL) and treated with hydroxylamine hydrochloride (319 mg, 4.60 mmol) and pyridine (2 mL). The reaction mixture was heated to 65° C. and stirred at that temperature for 30 min. After cooling to room temperature, the reaction mixture was concentrated under reduced pressure The residue was diluted with H2O (10 mL). The pH was adjusted to 5˜6 with 1 N HCl aqueous solution, and the mixture was partitioned with EtOAc (15 mL). The aqueous phase was extracted with EtOAc (2×15 mL). The combined organics were dried over anhydrous Na2SO4, filtered, and concentrated in vacuo to provide crude 4-(hydroxyimino)isochroman-6-yl ethyl(methyl)carbamate 277.3 (228 mg, 862 μmol) as a colorless oil which was used without further purification. 1H NMR (300 MHz, CDCl3) □ 7.67 (s, 1H), 7.10 (s, 2H), 4.78 (s, 2H), 4.67 (s, 2H), 3.51-3.38 (m, 2H), 3.03 (d, J=22.5 Hz, 3H), 1.29-1.22 (m, 3H).

Step 4.

A flask containing solution of Compound 277.3 (228 mg, 862 μmol) in MeOH (5 mL) was flushed with H2. Raney Ni (0.5 mL) was added and the reaction mixture was stirred at ambient temperature for 3 h under a H2 balloon. (Boc)2O (375 mg, 1.72 mmol) was added, and the mixture was stirred at ambient temperature for 5 h. The nickel catalyst was removed by filtration through a celite pad, and the filtrate was concentrated under reduced pressure. Purification by flash column chromatography (SiO2, gradient elution from 5% EtOAc/hexanes to 20% EtOAc afforded 4-((tert-butoxycarbonyl)amino)isochroman-6-yl ethyl(methyl)carbamate 277.4 (132 mg, 376 μmol) as a colorless oil. 1H NMR (300 MHz, CDCl3) δ 7.16 (s, 1H), 7.04-6.95 (m, 2H), 5.08 (d, J=8.7 Hz, 1H), 4.81-4.65 (m, 3H), 4.02 (dd, J=14.7, 2.7 Hz, 1H), 3.85 (dd, J=14.7, 3.0 Hz, 1H), 3.45-3.39 (m, 2H), 3.02 (d, J=21.9 Hz, 3H), 1.46 (s, 9H), 1.26-1.14 (m, 3H).

Step 5.

The compound of Example 277 was prepared as previously described in Example 1. MS (ESI): m/z 251 [M+H]. 1H NMR (300 MHz, CD3OD) δ 7.24-7.12 (m, 3H), 4.98 (d, J=15.3 Hz, 1H), 4.75 (d, J=15.3 Hz, 1H), 4.33 (s, 1H), 4.19 (d, J=12.9 Hz, 1H), 3.94 (dd, J=12.9, 2.1 Hz, 1H), 3.53-3.35 (m, 2H), 3.05 (d, J=31.8 Hz, 3H), 1.19 (dt, J=24.0, 7.2 Hz, 3H).

TABLE 11 Compounds were prepared as described in Example 277 using the appropriately substituted starting materials. Example Structure Characterization Data 278 MS (ESI): m/z 178 [M + H]. 1H NMR (300 MHz, CDCl3) δ 8.67 (brs, 3H), 7.43 (s, 1H), 7.12 (d, J = 8.1 Hz, 1H), 6.95 (d, J = 7.8 Hz, 1H), 5.04-4.98 (m, 1H), 4.30 (s, 1H), 4.10 (s, 2H), 2.30 (s, 3H), 1.45 (d, J = 6.6 Hz, 3H). 279 MS (ESI): m/z 265 [M + H]. 1H NMR (300 MHz, DMSO-d6) δ 8.30 (brs, 3H), 7.49 (d, J = 8.4 Hz, 1H), 7.08 (dd, J = 8.4, 2.4 Hz, 1H), 6.96 (d, J = 2.4 Hz, 1H), 4.85 (d, J = 15.9 Hz, 1H), 4.75 (d, J = 15.9 Hz, 1H), 4.25 (s, 1H), 3.99-3.92 (m, 1H), 3.44-3.26 (m, 2H), 2.95 (d, J = 36.0 Hz, 3H), 1.32 (d, J = 6.6 Hz, 3H), 1.12 (dt, J = 24.5, 7.2 Hz, 3H). 280 MS (ESI): m/z 265 [M + H]. 1H NMR (300 MHz, DMSO-d6) δ 8.57 (brs, 3H), 7.59 (d, J = 8.4 Hz, 1H), 7.10 (dd, J = 8.4, 2.1 Hz, 1H), 6.97 (d, J = 2.1 Hz, 1H), 4.73 (s, 2H), 4.18-4.13 (m, 2H), 344-3.26 (m, 2H), 2.95 (d, J = 36.0 Hz, 3H), 1.27 (d, J = 6.3 Hz, 3H), 1.13 (dt, J = 24.4, 6.9 Hz, 3H). 281 MS (ESI): m/z 265 [M + H]. 1H NMR (300 MHz, CD3OD) δ 7.26-7.16 (m, 3H), 4.99 (d, J = 15.6 Hz, 1H), 4.85 (d, J = 15.6 Hz, 1H), 4.28 (s, 1H), 4.05 (q, J = 6.6 Hz, 1H), 3.57-3.31 (m, 2H), 3.05 (d, J = 34.1 Hz, 3H), 1.41 (d, J = 6.6 Hz, 3H), 1.24 (dt, J = 23.7, 7.2 Hz, 3H). 282 MS (ESI): m/z 265 [M + H]. 1H NMR (300 MHz, CD3OD) δ 7.09-7.00 (m, 3H), 4.69 (s, 2H), 4.17- 4.04 (m, 2H), 3.40-3.32 (m, 2H), 2.89 (d, J = 34.8 Hz, 3H), 1.17 (d, J = 6.6 Hz, 3H), 1.09 (dt, J = 24.6, 7.2 Hz, 3H).

General Procedure for Chiral Separation of Racemic Compounds.

The chiral separations were carried out on the racemic amines or N-Boc protected amine intermediates using SFC (supercritical CO2 fluid chromatography) on a preparative Thar SFC-80 system using the specified column and co-solvent system. CO2 total flows were between 60 to 80 g/min with 100 bar back pressure. Additional examples were separated by normal phase preparatory HPLC using the specified column and solvent system.

The Faster Moving Enantiomer (FME) was the earlier eluting enantiomer and the Slower Moving Enantiomer (SME) was the later eluting enantiomer. Following isolation of the separated enantiomers, the Boc protecting groups were removed (if needed) and HCl salts were formed as previously described in Example 1. The compounds in Table 12 are all single enantiomers prepared by chiral separation of previously described racemates. For example, Examples 111 and 112 are the individual enantiomers of Example 12. Each example is a single enantiomer, but the absolute stereochemistry has not been conclusively proven.

TABLE 12 Compounds Prepared by Chiral Separation Example Separation Conditions Analytical Data SFC AD-H 20 × 250 mm, 5 μm 65:35 CO2/MeOH (0.1% NH4OH) FME MS (ESI): m/z 150 [M + H]. 1H NMR (400 MHz, CD3OD) δ 7.36-7.49 (m, 3H), 7.20 (d, J = 7.6 Hz, 1H), 4.95 (d, J = 15.2 Hz, 1H), 4.82 (d, J = 15.6 Hz, 1H), 4.38 (s, 1H), 4.26 (dd, J = 12.8, 1.6 Hz, 1H), 3.99 (dd, J = 12.8, 2.0 Hz, 1H) SFC AD-H 20 × 250 mm, 5 μm 65:35 CO2/MeOH (0.1% NH4OH) SME MS (ESI): m/z 150 [M + H]. 1H NMR (400 MHz, CD3OD) δ 7.36-7.49 (m, 3H), 7.20 (d, J = 7.6 Hz, 1H), 4.95 (d, J = 15.2 Hz, 1H), 4.82 (d, J = 15.6 Hz, 1H), 4.38 (s, 1H), 4.26 (dd, J = 12.8, 1.6 Hz, 1H,), 3.99 (dd, J = 12.8, 2.0 Hz, 1H) SFC AD 20 × 250 mm, 5 μm 75:25 CO2/MeOH (0.1% NH4OH) FME MS (ESI): m/z 168 [M + H]. 1H NMR (500 MHz, CD3OD) δ 7.28-7.18 (m, 3H), 4.94 (d, J = 15.0 Hz, 1H,), 4.78 (d, J = 15.5 Hz, 1H), 4.40 (s, 1H), 4.25 (dd, J = 12.5, 1.5 Hz, 1H), 3.98 (dd, J = 12.5, 2.5 Hz, 1H). SFC AD 20 × 250 mm, 5 μm 75:25 CO2/MeOH (0.1% NH4OH) SME MS (ESI): m/z 168 [M + H]. 1H NMR (500 MHz, CD3OD) δ 7.28-7.18 (m, 3H), 4.94 (d, J = 15.0 Hz, 1H), 4.78 (d, J = 15.5 Hz, 1H), 4.40 (s, 1H), 4.25 (dd, J = 12.5, 1.5 Hz, 1H), 3.98 (dd, J = 12.5, 2.5 Hz, 1H). SFC AD 20 × 250 mm, 5 μm 80:20 CO2/MeOH (0.5% NH3) FME MS (ESI): m/z 177 [M + H]. 1H NMR (500 MHz, CD3OD) δ 7.17 (d, J = 11.6 Hz, 3H), 4.82 (d, J = 14.6 Hz, 1H), 4.78 (d, J = 8.6 Hz, 1H), 4.74 (d, J = 14.6 Hz, 1H),4.21-4.14 (m, 1H), 4.07-4.02 (m, 1H), 2.40 (s, 3H), 2.19-2.28 (m, 2H). SFC AD 20 × 250 mm, 5 μm 80:20 CO2/MeOH (0.5% NH3) SME MS (ESI): m/z 177 [M + H]. 1H NMR (500 MHz, CD3OD) δ 7.17 (d, J = 11.4 Hz, 3H), 4.82 (d, J = 14.6 Hz, 1H), 4.78 (d, J = 8.4 Hz, 1H), 4.74 (d, J = 14.6 Hz, 1H), 4.22-4.14 (m, 1H), 4.07-4.02 (m, 1H), 2.40 (s, 3H), 2.30-2.15 (m, 2H). SFC AD-H 20 × 250 mm, 5 μm 60:30 CO2/MeOH (0.5% NH4OH) FME MS (ESI): m/z 183 [M − 16]. 1H-NMR of freebase (500 MHz, CDCl3) δ 8.17 (d, J = 8.5 Hz, 1H), 7.87 (d, J = 8.0 Hz, 1H), 7.76 (d, J = 8.5 Hz, 1H), 7.63-7.59 (m, 1H), 7.53- 7.50 (m, 1H), 7.12 (d, J = 8.5 Hz, 1H), 4.99- 4.88 (m, 2H), 4.38 (s, 1H), 4.27 (d, J = 11.5 Hz, 1H), 4.00-3.97 (dd, J = 11.0, 2.5 Hz, 1H), 1.90 (s, 2H). SFC AD-H 20 × 250 mm, 5 μm 60:40 CO2/MeOH (0.5% NH4OH) SME MS (ESI): m/z 183 [M − 16]. 1H-NMR of freebase (500 MHz, CDCl3) δ 8.16 (d, J = 9.0 Hz, 1H), 7.87 (d, J = 8.0 Hz, 1H), 7.76 (d, J = 8.5 Hz, 1H), 7.63-7.59 (m, 1H), 7.53- 7.50 (m, 1H), 7.12 (d, J = 8.5 Hz, 1H), 4.99- 4.88 (m, 2H), 4.39 (s, 1H), 4.28 (d, J = 11.5 Hz, 1H), 4.00-3.97 (dd, J = 11.0, 2.5 Hz, 1H), 1.92 (s, 2H). SFC AD 20 × 250 mm, 5 μm 60:30 CO2/MeOH (0.1% NH4OH) FME MS (ESI): m/z 218 [M + H]. 1H NMR (400 MHz, CD3OD) δ 7.64 (d, J = 8.4 Hz, 1H), 7.21 (d, J = 8.4 Hz, 1H), 4.96 (d, J = 15.9 Hz, 1H), 4.78 (d, J = 15.9 Hz, 1H), 4.67 (s, 1H), 4.32 (d, J = 13.0 Hz, 1H), 3.92 (dd, J = 13.0, 1.8 Hz, 1H). SFC AD 20 × 250 mm, 5 μm 60:40 CO2/MeOH (0.1% NH4OH) SME MS (ESI): m/z 218 [M + H]. 1H NMR (400 MHz, CD3OD) δ 7.64 (d, J = 8.4 Hz, 1H), 7.21 (d, J = 8.4 Hz, 1H), 4.96 (d, J = 15.9 Hz, 1H), 4.78 (d, J = 15.9 Hz, 1H), 4.66 (s, 1H), 4.32 (d, J = 13.0 Hz, 1H), 3.92 (dd, J = 13.0, 1.9 Hz, 1H). SFC AD 20 × 250 mm, 5 μm 75:25 CO2/MeOH (0.5% NH3) FME MS (ESI): m/z 180 [M + H]. 1H NMR (400 MHz, CD3OD) δ 7.11 (d, J = 8.5 Hz, 1H), 7.05 (d, J = 2.5 Hz, 1H), 7.00 (dd, J = 8.5, 2.6 Hz, 1H), 4.85 (d, J = 14.9 Hz, 1H), 4.73 (d, J = 14.9 Hz, 1H), 4.34 (s, 1H), 4.23 (dd, J = 12.8, 1.4 Hz, 1H), 3.95 (dd, J = 12.8, 2.3 Hz, 1H), 3.83 (s, 3H). SFC AD 20 × 250 mm, 5 μm 75:25 CO2/MeOH (0.5% NH3) SME MS (ESI): m/z 180 [M + H]. 1H NMR (400 MHz, CD3OD) δ 7.11 (d, J = 8.4 Hz, 1H), 7.05-6.96 (m, 2H), 4.85 (d, J = 14.9 Hz, 1H), 4.74 (d, J = 14.9 Hz, 1H), 4.32 (s, 1H), 4.22 (dd, J = 12.8, 2.0 Hz, 1H), 3.95 (dd, J = 12.8, 2.3 Hz, 1H), 3.83 (s, 3H). SFC AY 20 × 250 mm, 5 μm 90:10 CO2/MeOH (0.5% NH3) FME MS (ESI): m/z 164 [M + H]. 1H NMR (400 MHz, CD3OD) δ 7.28-7.24 (m, 2H), 7.07 (d, J = 8.0 Hz, 1H), 4.91-4.88 (m, 1H), 4.76 (d, J = 15.2 Hz, 1 H), 4.32 (s, 1H), 4.23 (d, J = 12.8 Hz, 1H), 3.95 (dd, J = 12.8, 2.4 Hz, 1H), 2.37 (s, 3H). SFC AY 20 × 250 mm, 5 μm 90:10 CO2/MeOH (0.5% NH3) SME MS (ESI): m/z 164 [M + H]. 1H NMR (400 MHz, CD3OD) δ 7.28-7.23 (m, 2H), 7.07 (d, J = 8.0 Hz, 1H), 4.92-4.88 (m, 1H), 4.75 (d, J = 15.2 Hz, 1 H), 4.32 (s, 1H), 4.23 (dd, J = 12.8, 1.2Hz, 1H), 3.95 (dd, J = 12.8, 2.4 Hz, 1H), 2.37 (s, 3H). HPLC AS-H 20 × 250, 10 □m 95:5 Hexanes/EtOH (0.1% DEA) FME MS (ESI): m/z 178 [M + H]. 1H NMR (500 MHz, CD3OD) δ 7.10 (d, J = 7.2 Hz, 2H), 4.89 (s, 1H), 4.68 (d, J = 15.5 Hz, 1H), 4.30 (s, 1H), 4.22 (d, J = 12.7 Hz, 1H), 3.92 (d, J = 12.6 Hz, 1H), 2.35 (s, 3H), 2.18 (s, 3H). HPLC AS-H 20 × 250, 10 □m 95:5 Hexanes/EtOH (0.1% DEA) SME MS (ESI): m/z 178 [M + H]. 1H NMR (500 MHz, CD3OD) δ 7.11 (d, J = 15.7 Hz, 2H), 4.89 (s, 1H), 4.67 (d, J = 15.5 Hz, 1H), 4.31 (s, 1H), 4.23 (d, J = 12.7 Hz, 1H), 3.92 (d, J = 12.6 Hz, 1H), 2.35 (s, 3H), 2.18 (s, 3H). SFC AD 20 × 250 mm, 5 μm 70:30 CO2/MeOH (0.5% NH3) SME MS (ESI): m/z 184.0 [M + H]. 1H NMR (400 MHz, CD3OD) δ 7.47-7.42 (m, 2H), 7.19 (dd, J = 6.0, 3.2 Hz, 1 H), 5.00-4.78 (m, 2 H), 4.63 (s, 1 H), 4.33 (dd, J = 12.8, 0.8 Hz, 1 H), 3.93 (dd, J = 13.2, 2.0 Hz, 1 H). SFC AD 20 × 250 mm, 5 μm 70:30 CO2/MeOH (0.5% NH3) FME MS (ESI): m/z 184.0 [M + H]. 1H NMR (400 MHz, CD3OD) δ 7.46-7.42(m, 2H), 7.18- 7.20 (m, 1 H), 4.99-4.78 (m, 2 H), 4.62 (s, 1 H), 4.34-4.31 (m, 1 H), 3.93 (dd, J = 12.8, 1.6 Hz, 1 H). SFC AY 20 × 250 mm, 5 μm 50:50 CO2/MeOH (0.5% NH3) FME MS (ESI): m/z 164.1 [M + H]. 1H NMR (400 MHz, DMSO-d6) δ 8.73 (br s, 3 H), 7.42 (d, J = 6.8 Hz, 1 H), 7.24-7.18 (m, 2 H), 4.79- 4.59 (m, 2H), 4.28 (s, 1 H), 4.22 (dd, J = 12.4, 2.0 Hz, 1H), 3.84 (dd, J = 12.4, 2.0 Hz, 1 H), 2.13 (s, 3 H). SFC AY 20 × 250 mm, 5 μm 50:50 CO2/MeOH (0.5% NH3) SME MS (ESI): m/z 164.1 [M + H]. 1H NMR (400 MHz, DMSO-d6) δ 8.69 (br s, 3 H), 7.42 (d, J = 6.8 Hz, 1 H), 7.24-7.18 (m, 2 H), 4.79- 4.59 (m, 2H), 4.28 (s, 1 H), 4.20 (dd, J = 12.4, 1.6 Hz, 1 H), 3.83 (dd, J = 12.4, 2.4 Hz, 1 H), 2.13 (s, 3 H). SFC OJ 20 × 250 mm, 5 μm 70:30 CO2/MeOH (0.5% NH3) FME MS (ESI): m/z 184 [M + H]. 1H NMR (400 MHz, DMSO-d6) δ 8.88 (br s, 3H), 7.64 (d, J = 7.6 Hz, 1H), 7.50-7.48 (m, 1H), 7.38 (t, J = 8.0 Hz, 1H), 4.82-4.61 (m, 2H), 4.36 (s, 1H), 4.28-4.25 (m, 1H), 3.88 (dd, J = 12.4, 2.4 Hz, 1H). SFC OJ 20 × 250 mm, 5 μm 70:30 CO2/MeOH (0.5% NH3) SME MS (ESI): m/z 184 [M + H]. 1H NMR (400 MHz, DMSO-d6) δ 8.87 (br s, 3H), 7.64 (d, J = 7.2 Hz, 1H), 7.48 (dd, J = 8.4, 0.8 Hz, 1H), 7.38 (t, J = 8.0 Hz, 1H), 4.82-4.61 (m, 2H), 4.37 (s, 1H), 4.28-4.25 (m, 1H), 3.88 (dd, J = 12.8, 2.4 Hz, 1H). SFC IC 30 × 250 mm, 5 μm 90:10 CO2/MeOH (0.1% NH4OH) FME MS (ESI): m/z 164 [M + H]. 1H NMR (400 MHz, DMSO-d6) δ 8.56 (br s, 3H), 7.25 (d, J = 7.6 Hz, 1H), 7.12 (d, J = 7.2 Hz, 1H), 6.97 (d, J = 8.0 Hz, 1H), 4.87-4.68 (m, 2H), 4.32 (s, 1H), 4.30 (d, J = 12.0 Hz, 1H), 3.75 (d, J = 11.2 Hz, 1H), 2.42 (s, 3H). SFC IC 30 × 250 mm, 5 μm 90:10 CO2/MeOH (0.1% NH4OH) SME MS (ESI): m/z 164 [M + H]. 1H NMR (400 MHz, DMSO-d6) δ 8.54 (br s, 3H), 7.26 (t, J = 7.6 Hz, 1H), 7.12 (d, J = 7.2 Hz, 1H), 6.97 (d, J = 7.6 Hz, 1H), 4.87-4.68 (m, 2H), 4.39 (s, 1H), 4.30 (d, J = 12.0 Hz, 1H), 3.78-3.75 (m, 1H), 2.42 (s, 3H). SFC AD 20 × 250 mm, 5 μm 55:45 CO2/MeOH (0.5% NH3) FME MS (ESI): m/z 178 [M + H]. 1H NMR (500 MHz, CD3OD) δ 7.22 (d, J = 8.0 Hz, 1H), 6.92 (d, J = 8.0 Hz, 1H), 4.93 (d, J = 15.0 Hz, 1H), 4.79 (d, J = 15.0 Hz, 1H), 4.58 (s, 1H), 4.30 (dd, J = 12.5, 1.0 Hz, 1H), 3.90 (d, J = 12.5 Hz, 1H), 2.34 (s, 3H), 2.33 (s, 3H). SFC AD 20 × 250 mm, 5 μm 55:45 CO2/MeOH (0.5% NH3) SME MS (ESI): m/z 178 [M + H]. 1H NMR (500 MHz, CD3OD) δ 7.23 (d, J = 7.5 Hz, 1H), 6.93 (d, J = 7.5 Hz, 1H), 4.93 (d, J = 15.0 Hz, 1H), 4.79 (d, J = 15.0 Hz, 1H), 4.61 (s, 1H), 4.30 (dd, J = 12.5, 1.0Hz, 1H), 3.91 (d, J = 12.5 Hz, 1H), 2.34 (s, 3H), 2.33 (s, 3H). SFC AY 4.6 × 250 mm, 5 μm 85:15 CO2/EtOH (0.1% NH3) FME MS (ESI): m/z 178 [M + H]. 1H NMR (500 MHz, CD3OD) δ 7.35 (d, J = 7.6 Hz, 1H), 7.25 (d, J = 7.5 Hz, 1H), 7.05 (d, J = 7.7 Hz, 1H), 5.00 (d, J = 15.6 Hz, 1H), 4.84 (d, J = 15.6 Hz, 1H), 4.55-4.41 (m, 2H), 3.90 (dd, J = 13.1, 1.3 Hz, 1H), 2.85 (s, 3H), 2.47 (s, 3H). SFC AY 4.6 × 250 mm, 5 μm 85:15 CO2/EtOH (0.1% NH3) SME MS (ESI): m/z 178 [M + H]. 1H NMR (500 MHz, CD3OD) δ 7.36 (t, J = 7.6 Hz, 1H), 7.25 (d, J = 7.5 Hz, 1H), 7.04 (d, J = 7.7 Hz, 1H), 4.99 (d, J = 15.6 Hz, 1H), 4.84 (d, J = 15.7 Hz, 1H), 4.53-4.42 (m, 2H), 3.90 (dd, J = 13.1, 1.2 Hz, 1H), 2.85 (s, 3H), 2.47 (s, 3H). HPLC AY-H 4.6 × 250 mm, 5 μm 90:10 Hexanes/EtOH (0.1% DEA) FME MS (ESI) m/z 194 [M + H]. 1H NMR (400 MHz, CD3OD) δ 7.14 (d, J = 8.5 Hz, 1H), 7.10-7.01 (m, 2H), 4.91 (d, J = 12.0 Hz, 4H), 4.76 (d, J = 15.0 Hz, 1H), 4.43 (dd, J = 13.3, 0.8 Hz, 1H), 4.25 (s, 1H), 3.91 (dd, J = 13.3, 1.9 Hz, 1H), 3.85 (s, 3H), 2.77 (s, 3H). HPLC AY-H 4.6 × 250 mm, 5 μm 90:10 Hexanes/EtOH (0.1% DEA) SME MS (ESI): m/z 194 [M + H]. 1H NMR (400 MHz, CD3OD) δ 7.14 (d, J = 8.5 Hz, 1H), 7.10-7.02 (m, 2H), 4.90 (d, J = 12.0 Hz, 1H), 4.76 (d, J = 15.0 Hz, 1H), 4.43 (dd, J = 13.2, 1.0 Hz, 1H), 4.25 (s, 1H), 3.91 (dd, J = 13.2, 1.9 Hz, 1H), 3.85 (s, 3H), 2.77 (s, 3H). HPLC AY-H 4.6 × 250 mm, 5 μm 90:10 Hexanes/EtOH (0.1% DEA) FME MS (ESI): m/z 178 [M + H]. 1H NMR (400 MHz, CD3OD) δ 7.32 (s, 1H), 7.28 (d, J = 8.0 Hz, 1H), 7.10 (d, J = 8.0 Hz, 1H), 4.93 (d, J = 15.2 Hz, 1H), 4.78 (d, J = 15.2 Hz, 1H), 4.43 (d, J = 12.8 Hz, 1H), 4.23 (s, 1H), 3.92 (dd, J = 13.2, 2.0 Hz, 1H), 2.76 (s, 3H), 2.38 (s, 3H). HPLC AY-H 4.6 × 250 mm, 5 μm 90:10 Hexanes/EtOH (0.1% DEA) SME MS (ESI): m/z 178 [M + H]. 1H NMR (400 MHz, CD3OD) δ 7.32 (s, 1H), 7.28 (d, J = 8.0 Hz, 1H), 7.10 (d, J = 8.0 Hz, 1H), 4.93 (d, J = 15.2 Hz, 1H), 4.78 (d, J = 15.2 Hz, 1H), 4.43 (d, J = 12.8 Hz, 1H), 4.23 (s, 1H), 3.92 (dd, J = 13.2, 2.0 Hz, 1H), 2.76 (s, 3H), 2.38 (s, 3H). SFC AY-H 4.6 × 250 mm, 5 μm 80:20 CO2/EtOH (1% NH3) FME MS (ESI): m/z 192 [M +H]. 1H NMR (400 MHz, CD3OD) δ 7.25 (d, J = 7.8 Hz, 1H), 6.93 (d, J = 7.8 Hz, 1H), 4.94 (d, J = 15.4 Hz, 1H), 4.80 (d, J = 15.4 Hz, 1H), 4.53~ 4.43 (m, 2H), 3.91~3.82 (m, 1H), 2.83 (s, 3H), 2.33 (s, 6H). SFC AY-H 4.6 × 250 mm, 5 μm 80:20 CO2/EtOH (1% NH3) SME MS (ESI): m/z 192 [M +H]. 1H NMR (400 MHz, CD3OD) δ 7.27 (d, J = 8.0 Hz, 1H), 6.94 (d, J = 8.0 Hz, 1H), 4.94 (d, J = 15.4 Hz, 1H), 4.80 (d, J = 15.3 Hz, 1H), 4.48 (dd, J = 15.2, 1.9 Hz, 2H), 3.86 (dd, J = 12.9, 0.9 Hz, 1H), 2.82 (s, 3H), 2.33 (s, 6H). HPLC AY-H 4.6 × 250 mm, 5 μm 80:20 Hexanes/EtOH (0.1% DEA) FME MS (ESI): m/z 198 [M + H]. 1H NMR (500 MHz, CD3OD) δ 7.36 (t, J = 7.6 Hz, 1H), 7.25 (d, J = 7.5 Hz, 1H), 7.04 (d, J = 7.7 Hz, 1H), 4.99 (d, J = 15.6 Hz, 1H), 4.84 (d, J = 15.7 Hz, 1H), 4.53-4.42 (m, 2H), 3.90 (dd, J = 13.1, 1.2 Hz, 1H), 2.85 (s, 3H), 2.47 (s, 3H). HPLC AY-H 4.6 × 250 mm, 5 μm 80:20 Hexanes/EtOH (0.1% DEA) SME MS (ESI): m/z 198 [M + H]. 1H NMR (500 MHz, CD3OD) δ 7.53-7.47 (m, 2H), 7.22 (dd, J = 8.6, 6.6 Hz, 1H), 5.01 (d, J = 15.9 Hz, 1H), 4.83 (d, J = 15.9 Hz, 1H), 4.59- 4.46 (m, 2H), 3.92 (dd, J = 13.4, 1.6 Hz, 1H), 2.88 (s, 3H). HPLC AY-H 4.6 × 250 mm, 5 μm 90:10 Hexanes/EtOH (0.1% DEA) FME MS (ESI): m/z 232 [M + H]. 1H NMR (400 MHz, CD3OD) δ 7.67 (d, J = 8.8 Hz, 1H), 7.23 (d, J = 8.4 Hz, 1H), 4.99 (d, J = 16.0 Hz, 1H), 4.79 (d, J = 16.4 Hz, 1H), 4.59 (s, 1H), 4.52 (d, J = 13.6 Hz, 1H), 3.89 (dd, J = 12.8, 1.2 Hz, 1H), 2.89 (s, 3H). HPLC AY-H 4.6 × 250 mm, 5 μm 90:10 Hexanes/EtOH (0.1% DEA) SME MS (ESI): m/z 232 [M + H]. 1H NMR (400 MHz, CD3OD) δ 7.67 (d, J = 8.8 Hz, 1H), 7.21 (d, J = 8.4 Hz, 1H), 4.99 (d, J = 16.0 Hz, 1H), 4.79 (d, J = 16.4 Hz, 1H), 4.59 (s, 1H), 4.52 (d, J = 13.6 Hz, 1H), 3.89 (dd, J = 12.8, 1.2 Hz, 1H), 2.89 (s, 3H). SFC AY 20 × 250 mm, 5 μm 80:20 CO2/EtOH (0.2% NH3) FME MS (ESI): m/z 198 [M + H]. 1H NMR (400 MHz, CD3OD) δ 7.52 (dd, J = 13.3, 8.0 Hz, 2H), 7.42 (t, J = 7.8 Hz, 1H), 5.03 (d, J = 16.3 Hz, 1H), 4.74 (d, J = 16.3 Hz, 1H), 4.46 (d, J = 13.2 Hz, 1H), 4.33 (s, 1H), 3.93 (dd, J = 13.2, 1.5 Hz, 1H), 2.79 (s, 3H). SFC AY-H 20 × 250 mm, 5 μm 80:20 CO2/EtOH (0.2% NH3) SME MS (ESI): m/z 198 [M + H]. 1H NMR (400 MHz, CD3OD) δ 7.41 (dd, J = 16.0, 6.6 Hz, 2H), 7.31 (t, J = 7.8 Hz, 1H), 4.92 (d, J = 16.3 Hz, 1H), 4.63 (d, J = 16.3 Hz, 1H), 4.35 (d, J = 13.3 Hz, 1H), 4.22 (s, 1H), 3.82 (dd, J = 13.2, 1.5 Hz, 1H), 2.68 (s, 3H). SFC OD 4.6 × 250 mm, 5 μm 90:10 CO2/MeOH (0.2% NH3) FME MS (ESI): m/z 164 [M + H]. 1H NMR (400 MHz, CD3OD) δ 7.47 (m, 1H), 7.43 (ddd, J = 7.2, 7.2, 0.8 Hz, 1H), 7.37 (dd, J = 8.0, 8.0 Hz, 1H), 7.20 (d, J = 7.2 Hz, 1H), 4.86 (s, 2H), 4.30 (m, 1H), 4.21 (s, 1H), 1.33 (d, J = 6.8 Hz, 3H). SFC OD 4.6 × 250 mm, 5 μm 90:10 CO2/MeOH (0.2% NH3) SME MS (ESI): m/z 164 [M + H]. 1H NMR (400 MHz, CD3OD) δ 7.48 (m, 1H), 7.42 (ddd, J = 7.6, 7.6, 1.2 Hz, 1H), 7.37 (dd, J = 7.6, 7.6, Hz, 1H), 7.20 (d, J = 7.2 Hz, 1H), 4.86 (s, 2H), 4.31 (m, 1H), 4.22 (s, 1H), 1.33 (d, J = 6.8 Hz, 3H). SFC IC 20 × 250 mm, 5 μm 85:15 CO2/MeOH (0.5% NH3) FME MS (ESI): m/z 164 [M + H]. 1H NMR (400 MHz, CD3OD) δ 7.45-7.34 (m, 3H), 7.20 (d, J = 7.2 Hz, 1H), 4.98 (d, J = 15.6 Hz, 1H), 4.90-4.86 (m, 1H), 4.28 (s, 1H), 4.08-4.03 (m, 1H), 1.42 (d, J = 6.4 Hz, 3H). SFC IC 20 × 250 mm, 5 μm 85:15 CO2/MeOH (0.5% NH3) SME MS (ESI): m/z 164 [M + H]. 1H NMR (400 MHz, CD3OD) δ 7.47-7.34 (m, 3H), 7.20 (d, J = 7.6 Hz, 1H), 4.98 (d, J = 15.6 Hz, 1H), 4.90-4.86 (m, 1H), 4.29 (s, 1H), 4.07-4.03 (m, 1H), 1.42 (d, J = 6.4 Hz, 3H). SFC AD 4.6 × 250 mm, 5 μm 85:15 CO2/MeOH (0.2% NH3) FME MS (ESI): m/z 178 [M + H]. 1H NMR (500 MHz, CD3OD) δ 7.27 (s, 1H), 7.26 (d, J = 8.2 Hz, 1H), 7.09 (d, J = 7.7 Hz, 1H), 4.83 (d, J = 2.2 Hz, 2H), 4.27 (qd, J = 6.7, 2.6 Hz, 1H), 4.15 (d, J = 2.4 Hz, 1H), 2.39 (s, 3H), 1.33 (d, J = 6.7 Hz, 3H). SFC AD 4.6 × 250 mm, 5 μm 85:15 CO2/MeOH (0.2% NH3) SME MS (ESI): m/z 178 [M + H]. 1H NMR (500 MHz, CD3OD) δ 7.27 (s, 1H), 7.26 (d, J = 8.6 Hz, 1H), 7.09 (d, J = 7.7 Hz, 1H), 4.83 (s, 2H), 4.28 (qd, J = 6.6, 2.6 Hz, 1H), 4.15 (d, J = 2.4 Hz, 1H), 2.39 (s, 3H), 1.33 (d, J = 6.7 Hz, 3H). HPLC AY-H 4.6 × 250 mm, 5 μm 90:10 Hexanes/EtOH (0.1% Et2NH) FME MS (ESI): m/z 178 [M + H]. 1H NMR (400 MHz, CD3OD) δ 7.26 (s, 1H), 7.24 (d, J = 8.0 Hz, 1H), 7.08 (d, J = 8.0 Hz, 1H), 4.93 (d, J = 12.0 Hz, 1H), 4.84 (d, J = 15.3 Hz, 1H), 4.22 (s, 1H), 4.03 (dd, J = 6.6, 1.6 Hz, 1H), 2.38 (s, 3H), 1.41 (d, J = 6.6 Hz, 3H). HPLC AY-H 4.6 × 250 mm, 5 μm 90:10 Hexanes/EtOH (0.1% Et2NH) SME MS (ESI): m/z 178 [M + H]. 1H NMR (400 MHz, CD3OD) δ 7.26 (s, 1H), 7.25 (d, J = 8.1 Hz, 1H), 7.08 (d, J = 7.7 Hz, 1H), 4.94 (d, J = 12.0 Hz, 1H), 4.84 (d, J = 15.4 Hz, 1H), 4.23 (s, 1H), 4.03 (dd, J = 6.6, 1.6 Hz, 1H), 2.38 (s, 3H), 1.41 (d, J = 6.6 Hz, 3H). SFC IC 20 × 250 mm, 5 μm 80:20 CO2/MeOH (0.2% NH3) FME MS (ESI): m/z 198 [M + H]. 1H NMR (400 MHz, CD3OD) δ 7.47-7.44 (m, 2H), 7.27- 7.17 (m, 1H), 4.88 (s, 2H), 4.48-4.43 (m, 2H), 1.28 (d, J = 6.8 Hz, 3H). SFC IC 20 × 250 mm, 5 μm 80:20 CO2/MeOH (0.2% NH3) SME MS (ESI): m/z 198 [M + H]. 1H NMR (400 MHz, CD3OD) δ 7.52-7.41 (m, 2H), 7.25- 7.16 (m, 1H), 4.88 (s, 2H), 4.53-4.39 (m, 2H), 1.28 (d, J = 6.8 Hz, 3H). SFC AY 30 × 250 mm, 5 μm 80:20 CO2/MeOH (0.1% NH4OH) FME MS (ESI): m/z 161 [M − 16]. 1H NMR (400 MHz, CD3OD) δ 8.48 (bs, 3 H), 7.10 (dd, J1 = 6.4 Hz, J2 =20 Hz, 2 H), 4.80 (d, J = 12.4 Hz, 1 H), 4.61 (d, J = 12.8 Hz, 1 H), 4.39 (s, 1 H), 4.27 (d, J = 9.6 Hz, 1 H), 3.73 (d, J = 9.6 Hz, 1 H), 2.38 (s, 3 H), 2.10 (s, 3 H). SFC AY 30 × 250 mm, 5 μm 80:20 CO2/MeOH (0.1% NH4OH) SME MS (ESI): m/z 161 [M − 16]. 1H NMR (400 MHz, CD3OD) δ 8.52 (bs, 3 H), 7.09 (m, 2 H), 4.81 (d, J = 13.2 Hz, 1 H), 4.61 (d, J = 12.4 Hz, 1 H), 4.39 (d, J = 2.8 Hz, 1 H), 4.28 (d, J = 10.0 Hz, 1 H), 3.72 (d, J = 9.6 Hz, 1 H), 2.39 (s, 3 H), 2.09 (s, 3 H). SFC IC 4.6 × 250 mm, 5 μm 85:15 CO2/MeOH (0.2% NH3) FME MS (ESI): m/z 178 [M + H]. 1H NMR (400 MHz, CD3OD) δ 7.34 (t, J = 7.6 Hz, 1H), 7.23 (d, J = 7.5 Hz, 1H), 7.03 (d, J = 7.7 Hz, 1H), 4.88 (s, 2H), 4.49-4.35 (m, 2H), 2.45 (s, 3H), 1.27 (d, J = 6.7 Hz, 3H). SFC IC 4.6 × 250 mm, 5 μm 85:15 CO2/MeOH (0.2% NH3) SME MS (ESI): m/z 178 [M + H]. 1H NMR (400 MHz, CD3OD) δ 7.34 (t, J = 7.6 Hz, 1H), 7.23 (d, J = 7.5 Hz, 1H), 7.03 (d, J = 7.7 Hz, 1H), 4.88 (s, 2H), 4.48-4.37 (m, 2H), 2.46 (s, 3H), 1.27 (d, J = 6.7 Hz, 3H). HPLC AY-H 20 × 250 mm, 5 μm 90:10 Hexanes/EtOH (0.1% Et2NH) FME MS (ESI): m/z 178 [M + H]. 1H NMR (400 MHz, CD3OD) δ 7.32 (t, J = 7.6 Hz, 1H), 7.22 (d, J = 7.5 Hz, 1H), 7.02 (d, J = 7.7 Hz, 1H), 4.99 (d, J = 15.6 Hz, 1H), 4.87 (s, 1H), 4.44 (s, 1H), 4.08-3.99 (m, 1H), 2.45 (s, 3H), 1.44 (d, J = 6.6 Hz, 3H). HPLC AY-H 20 × 250 mm, 5 μm 90:10 Hexanes/EtOH (0.1% Et2NH) SME MS (ESI): m/z 178 [M + H]. 1H NMR (400 MHz, CD3OD) δ 7.32 (t, J = 7.6 Hz, 1H), 7.22 (d, J = 7.5 Hz, 1H), 7.02 (d, J = 7.6 Hz, 1H), 4.99 (d, J = 15.6 Hz, 1H), 4.87 (s, 1H), 4.43 (s, 1H), 4.03 (d, J = 6.6 Hz, 1H), 2.44 (s, 3H), 1.43 (d, J = 6.6 Hz, 3H). SFC OD 20 × 250 mm, 5 μm 85:15 CO2/MeOH (0.2% NH3) FME MS (ESI): m/z 194 [M + H]. 1H NMR (400 MHz, CD3OD) δ 7.13 (d, J = 8.2 Hz, 1H), 7.04-7.01 (m, 2H), 4.86-4.75 (m, 2H), 4.25 (tt, J = 6.7, 3.3 Hz, 1H), 4.17 (d, J = 2.3 Hz, 1H), 3.85 (s, 3H), 1.34 (d, J = 6.7 Hz, 3H). SFC OD 20 × 250 mm, 5 μm 85:15 CO2/MeOH (0.2% NH3) SME MS (ESI): m/z 194 [M + H]. 1H NMR (400 MHz, CD3OD) δ 7.13 (d, J = 9.3 Hz, 1H), 7.04-7.01 (m, 2H), 4.81 (d, J = 1.7 Hz, 2H), 4.25 (dd, J = 6.6, 2.4 Hz, 1H), 4.17 (s, 1H), 3.85 (s, 3H), 1.34 (d, J = 6.7 Hz, 3H). SFC AD 4.6 × 250 mm, 5 μm 90:10 CO2/MeOH (0.2% NH3) FME MS (ESI): m/z 194 [M + H]. 1H NMR (400 MHz, CD3OD) δ 7.11 (s, 1H), 7.02 (d, J = 6.4 Hz, 2H), 4.90 (s, 1H), 4.84 (s, 1H), 4.24 (s, 1H), 4.03 (s, 1H), 1.41 (d, J = 6.6 Hz, 3H). SFC AD 4.6 × 250 mm, 5 μm 90:10 CO2/MeOH (0.2% NH3) SME MS (ESI): m/z 194 [M + H]. 1H NMR (400 MHz, CD3OD) δ 7.12 (d, J = 9.2 Hz, 1H), 7.05-6.99 (m, 2H), 4.90 (s, 1H), 4.82 (d, J = 15.0 Hz, 1H), 4.24 (s, 1H), 4.03 (dd, J = 6.6, 1.6 Hz, 1H), 3.85 (s, 3H), 1.41 (d, J = 6.6 Hz, 3H). SFC IC 30 × 250 mm, 5 μm 80:20 CO2/MeOH (0.1% NH4OH) FME MS (ESI): m/z 198 [M + H]. 1H NMR (400 MHz, CD3OD) δ 7.39 (d, J = 8.0 Hz, 1 H), 7.27 (d, J = 8.4 Hz, 1 H), 5.04 (d, J = 16.0 Hz, 1 H), 4.72 (d, J = 16.4 Hz, 1 H), 4.59- 4.58 (m, 1 H), 4.35-4.30 (m, 1 H), 3.90 (dd, J = 12.8, 1.2 Hz, 1 H), 2.48-2.47 (m, 3 H). SFC IC 30 × 250 mm, 5 μm 80:20 CO2/MeOH (0.1% NH4OH) SME MS (ESI): m/z 198 [M + H]. 1H NMR (400 MHz, CD3OD) δ 7.39 (d, J = 8.0 Hz, 1 H), 7.26 (d, J = 8.4 Hz, 1 H), 5.04 (d, J = 16.4 Hz, 1 H), 4.71 (d, J = 16.0 Hz, 1 H), 4.58- 4.56 (m, 1 H), 4.33-4.29 (m, 1 H), 3.90 (dd, J = 12.4, 1.2 Hz, 1 H), 2.47-2.46 (m, 3 H). SFC AD-H 20 × 250 mm, 5 μm 90:10 CO2/MeOH (0.2% NH3) FME MS (ESI): m/z 175 [M − 16]. 1H NMR (400 MHz, CD3OD) δ 7.29 (s, 1H), 7.26-7.24 (d, J = 8.0 Hz, 1H), 7.09-7.07 (d, J = 7.9 Hz, 1H), 4.80 (s, 2H), 4.21 (s, 1H), 4.03-3.99 (m, 1H), 2.39 (s, 3H), 1.66-1.56 (m, 2H), 1.10-1.06 (t, J = 7.4 Hz, 3H). SFC AD-H 20 × 250 mm, 5 μm 90:10 CO2/MeOH (0.2% NH3) SME MS (ESI): m/z 175 [M − 16]. 1H NMR (400 MHz, CD3OD) δ 7.30 (s, 1H), 7.26-7.24 (d, J = 8.0 Hz, 1H), 7.09-7.07 (d, J = 7.9 Hz, 1H), 4.80 (s, 2H), 4.21 (s, 1H), 4.04-4.00 (m, 1H), 2.39 (s, 3H), 1.66-1.57 (m, 2H), 1.10-1.06 (t, J = 7.4 Hz, 3H). HPLC AY-H 50 × 250 mm, 5 μm 90:10 Hexanes/EtOH (0.1% Et2NH) FME MS (ESI): m/z 192 [M + H]. 1H NMR (400 MHz, CD3OD) δ 7.27-7.25 (m, 2H), 7.09 (d, J = 7.6 Hz, 1H), 4.98 (d, J = 15.2 Hz, 1H), 4.83 (d, J = 15.6 Hz, 1H), 4.29 (s, 1H), 3.75- 3.72 (m, 1H), 2.39 (s, 3H), 1.76-1.66 (m, 2H), 1.16 (t, J = 7.2 Hz, 3H). HPLC AY-H 50 × 250 mm, 5 μm 90:10 Hexanes/EtOH (0.1% Et2NH) SME MS (ESI): m/z 192 [M + H]. 1H NMR (400 MHz, CD3OD) δ 7.27-7.25 (m, 2H), 7.09 (d, J = 7.6 Hz, 1H), 4.98 (d, J = 15.6 Hz, 1H), 4.83 (d, J = 15.2 Hz, 1H), 4.29 (s, 1H), 3.75-3.72 (m, 1H), 2.39 (s, 3H), 1.76-1.66 (m, 2H), 1.14 (t, J = 7.6 Hz, 3H). SFC IC 30 × 250 mm, 5 μm 90:10 CO2/MeOH (0.1% NH4OH) FME MS (ESI): m/z 196 [M + H]. 1H NMR (400 MHz, CD3OD) δ 7.30-7.20 (m, 3H), 4.82 (s, 2H), 4.29 (s, 1H), 4.03-4.00 (m, 1H), 1.70- 1.56 (m, 2H), 1.09 (t, J = 7.6 Hz, 3H). SFC IC 30 × 250 mm, 5 μm 90:10 CO2/MeOH (0.1% NH4OH) SME MS (ESI): m/z 196 [M + H]. 1H NMR (400 MHz, CD3OD) δ 7.27-7.18 (m, 3H), 4.82 (s, 2H), ), 4.27 (d, J = 2.0 Hz, 1H), 4.02-3.98 (m, 1H), 1.70-1.54 (m, 2H), 1.09 (t, J = 7.6 Hz, 3H). SFC IC 30 × 250 mm 5 μm 90:10 CO2/MeOH (0.1% NH4OH) FME MS (ESI): m/z 182 [M + H]. 1H NMR (400 MHz, CD3OD) δ 7.26-7.20 (m, 3H), 4.84 (s, 2H), 4.27-4.22 (m, 2H), 1.33 (d, J = 6.8 Hz, 3H). SFC IC 30 × 250 mm, 5 μm 90:10 CO2/MeOH (0.1% NH4OH) SME MS (ESI): m/z 196 [M + H]. 1H NMR (400 MHz, CD3OD) δ 7.26-7.19 (m, 3H), 4.84 (s, 2H), 4.28-4.22 (m, 2H), 1.33 (d, J = 6.8 Hz, 2H). SFC IC 20 × 250 mm, 5 μm 85:15 CO2/MeOH (0.2% NH3) FME MS (ESI): m/z 182 [M + H]. 1H NMR (400 MHz, CD3OD) δ 7.27-7.17 (m, 3 H), 4.96 (d, J = 15.6 Hz, 1 H), 4.85 (d, J = 15.6 Hz, 1 H), 4.32 (s, 1 H), 4.07-4.02 (m, 1 H), 1.42 (d, J = 6.4 Hz, 3 H). SFC IC 20 × 250 mm, 5 μm 85:15 CO2/MeOH (0.2% NH3) SME MS (ESI): m/z 182 [M + H]. 1H NMR (400 MHz, CD3OD) δ 7.27-7.17 (m, 3 H), 4.96 (d, J = 15.2 Hz, 1 H), 4.85 (d, J = 15.6 Hz, 1 H), 4.32 (s, 1 H), 4.07-4.02 (m, 1 H), 1.42 (d, J = 6.4 Hz, 3 H). SFC IC 30 × 250 mm, 5 μm 90:10 CO2/MeOH (0.1% NH4OH) FME MS (ESI): m/z 196 [M + H]. 1H NMR (400 MHz, CD3OD) δ 7.26-7.18 (m, 3H), 5.03 (d, J = 15.2 Hz, 1H), 4.86-4.83 (d, J = 15.2 Hz, 1H), 4.37 (s, 1H), 3.77-3.73 (m, 1H), 1.79- 1.65 (m, 2H), 1.16-1.12 (t, J = 7.6 Hz, 3H). SFC IC 30 × 250 mm, 5 μm 90:10 CO2/MeOH (0.1% NH4OH) SME MS (ESI): m/z 196 [M + H]. 1H NMR (400 MHz, CD3OD) δ 7.26-7.18 (m, 3H), 5.03 (d, J = 15.2 Hz, 1H), 4.86-4.83 (d, J = 15.2 Hz, 1H), 4.37 (s, 1H), 3.77-3.73 (m, 1H), 1.79- 1.65 (m, 2H), 1.16-1.12 (t, J = 7.6 Hz, 3H). SFC AY-H 4.7 × 100 mm, 5 μm EtOH (1% NH3) FME MS (ESI): m/z 161 [M − 16]. 1H NMR (400 MHz, CD3OD) δ 7.33 (d, J = 8.0 Hz, 1H), 7.17 (d, J = 8.0 Hz, 1H), 7.01 (s, 1H), 4.95- 4.82 (m, 2H), 4.23 (s, 1H), 4.05-4.04 (m, 1H), 2.35 (s, 3H), 1.41 (d, J = 6.8 Hz, 3H). SFC AY-H 4.7 × 100 mm, 5 μm EtOH (1% NH3) SME MS (ESI): m/z 161 [M − 16]. 1H NMR (400 MHz, CD3OD) δ 7.33 (d, J = 8.0 Hz, 1H), 7.17 (d, J = 8.0 Hz, 1H), 7.01 (s, 1H), 4.95- 4.82 (m, 2H), 4.23 (s, 1H), 4.05-4.04 (m, 1H), 2.35 (s, 3H), 1.41 (d, J = 6.8 Hz, 3H). HPLC AY 20 × 250 mm, 10 μm 90:10 Hexanes/EtOH (0.1% Et2NH) FME MS (ESI): m/z 178 [M + H]. 1H NMR (400 MHz, CD3OD) δ 7.47-7.42 (m, 2H), 7.38 (t, J = 7.6 Hz, 1H), 7.22 (d, J = 7.6 Hz, 1H), 5.05 (d, J = 15.6 Hz, 1H), 4.90 (d, J = 15.6 Hz, 1H), 4.35 (s, 1H), 3.78-3.75 (m, 1H), 1.79-1.66 (m, 2H), 1.16 (t, J = 7.2 Hz, 3H). HPLC AY 20 × 250 mm, 10 μm 90:10 Hexanes/EtOH (0.1% Et2NH) SME MS (ESI): m/z 178 [M + H]. 1H NMR (400 MHz, CD3OD) δ 7.48-7.42(m, 2H), 7.38 (t, J = 7.2 Hz, 1H), 7.22 (d, J = 7.6 Hz, 1H), 5.05 (d, J = 15.2 Hz, 1H), 4.90 (d, J = 15.2 Hz, 1H), 4.36 (s, 1H), 3.78-3.75 (m, 1H), 1.78-1.69 (m, 2H), 1.16 (t, J = 7.2 Hz, 3H). SFC OD-H 20 × 250 mm 5 μm 90:10 CO2/MeOH (1% NH3) FME MS (ESI): m/z 161 [M − 16]. 1H NMR (400 MHz, CD3OD) δ 7.48-7.36 (m, 3H), 7.21- 7.19 (d, J = 7.5 Hz, 1H), 4.85 (s, 2H), 4.27 (d, J = 2.1 Hz, 1H), 4.05-4.01 (m, 1H), 1.67- 1.56 (m, 2H), 1.09-1.07 (t, J = 7.4 Hz, 3H). SFC OD-H 20 × 250 mm, 5 μm 90:10 CO2/MeOH (1% NH3) SME MS (ESI): m/z 161 [M − 16]. 1H NMR (400 MHz, CD3OD) δ 7.46-7.36 (m, 3H), 7.21- 7.19 (d, J = 7.5 Hz, 1H), 4.85 (s, 2H), 4.25 (d, J = 2.1 Hz, 1H), 4.04-4.00 (m, 1H), 1.66- 1.55 (m, 2H), 1.11-1.07 (t, J = 7.4 Hz, 3H) SFC OZ 20 × 250 mm, 5 μm 75:25 CO2/EtOH (1% NH3) FME MS (ESI): m/z 178 [M +H]+. 1H NMR (400 MHz, CD3OD) δ 7.32 (d, J = 7.6 Hz, 1H), 7.29 (d, J = 8.0 Hz, 1H),7.03 (s, 1H), 4.87 (s, 2H), 4.28-4.23 (m, 1H), 4.15 (d, J = 2.0 Hz, 1H), 2.37 (s, 3H), 1.32 (d, J = 2.8 Hz, 3H). SFC OZ 20 × 250 mm, 5 μm 75:25 CO2/EtOH (1% NH3) SME MS (ESI): m/z 178 [M + H]+. 1H NMR (400 MHz, CD3OD) δ d 7.35 (d, J = 6.0 Hz, 1H), 7.29 (d, J = 8.0 Hz, 1H), 7.03 (s, 1H), 4.88 (s, 2H), 4.30-4.26 (m, 1H), 4.16 (s, 1H), 2.37 (s, 3H), 1.32 (d, J = 2.4 Hz, 3H). HPLC AY-H 4.6 × 250 mm, 5 μm 90:10 Hexanes/EtOH (0.1% Et2NH) FME MS (ESI): m/z 178 [M + H]+. 1H NMR (400 MHz, D2O) δ 7.22-7.21 (m, 2H), 7.19-7.16 (m, 1H), 5.15 (q, J = 6.8 Hz, 1H), 4.34-4.31 (m, 2H), 3.95 (dd, J = 14.0, 2.0 Hz, 1H), 2.17 (s, 3H), 1.40 (d, J = 7.8 Hz, 3H). HPLC AY-H 4.6 × 250 mm, 5 μm 90:10 Hexanes/EtOH (0.1% Et2NH) SME MS (ESI): m/z 178 [M + H]+. 1H NMR (400 MHz, D2O) δ 7.22-7.21 (m, 2H), 7.19-7.16 (m, 1H), 5.15 (q, J = 6.8 Hz, 1H), 4.34-4.31 (m, 2H), 3.95 (dd, J = 14.0, 2.0 Hz, 1H), 2.17 (s, 3H), 1.40 (d, J = 7.8 Hz, 3H).

Chiral Separation of tert-butyl (2,4,7,8,9,10-hexahydro-1H-7,10-methanobenzo-[f]isochromen-1-yl)carbamate

As previously described in the General Procedure for Chiral Separation above compound Boc-51 (prepared as previously described in Example 51) was separated into 3 peaks consisting of a mixture of enantiomers (P1 and P4) and the individual enantiomers P2 and P3 using an AD 20×250 mm, 5 um column and a mobile phase of 85:15 CO2/MeOH/0.5% NH3. The mixture of P1 and P4 was further separated by SFC into enantiomers P1 and P4 using a Whelk 20×250 mm, 5 um column and a mobile phase of 85:15 CO2/MeOH (0.5% NH3).

The individual Boc-protected enantiomers were deprotected and converted to the corresponding HCl salts as previously described in Example 1.

Example 161

FME on AD column, MS (ESI): m/z 216 [M+H]. 1H NMR (400 MHz, CD3OD): δ 7.22 (d, J=7.6 Hz, 1H), 6.87 (d, J=7.6 Hz, 1H), 4.89 (d, J=15.2 Hz, 1H), 4.79 (d, J=15.2 Hz, 1H), 4.56 (s, 1H), 4.32 (dd, J=12.8, 1.2 Hz, 1H), 3.91 (d, J=12.8, 2.0 Hz, 1H), 3.61 (s, 1H), 3.41 (d, J=2.0 Hz, 1H), 2.06-2.01 (m, 2H), 1.73-1.70 (m, 1H), 1.64-1.61 (m, 1H), 1.20-1.15 (m, 2H).

Example 162

SME on AD column, MS (ESI): m/z 216 [M+H]. 1H NMR (400 MHz, CD3OD): δ 7.20 (d, J=7.6 Hz, 1H), 6.87 (d, J=7.6 Hz, 1H), 4.94 (d, J=15.6 Hz, 1H), 4.77 (d, J=15.2 Hz, 1H), 4.43 (s, 1H), 4.25 (dd, J=12.8, 0.8 Hz, 1H), 3.91 (dd, J=12.8, 2.0 Hz, 1H), 3.66 (s, 1H), 3.39 (s, 1H), 1.96-1.99 (m, 2H), 1.84-1.81 (m, 1H), 1.65-1.62 (m, 1H), 1.10-1.06 (m, 2H).

Example 163

FME on Whelk column, MS (ESI): m/z 216 [M+H]. 1H NMR (400 MHz, CD3OD): δ 7.20 (d, J=7.6 Hz, 1H), 6.87 (d, J=7.6 Hz, 1H), 4.94 (d, J=15.6 Hz, 1H), 4.77 (d, J=15.2 Hz, 1H), 4.43 (s, 1H), 4.25 (dd, J=12.8, 0.8 Hz, 1H), 3.91 (dd, J=12.8, 2.0 Hz, 1H), 3.66 (s, 1H), 3.39 (s, 1H), 1.99-1.96 (m, 2H), 1.84-1.81 (m, 1H), 1.65-1.62 (m, 1H), 1.10-1.06 (m, 2H).

Example 164

SME on Whelk column, MS (ESI): m/z 216 [M+H]. 1H NMR (400 MHz, CD3OD): δ 7.22 (d, J=7.6 Hz, 1H), 6.87 (d, J=7.6 Hz, 1H), 4.89 (d, J=15.2 Hz, 1H), 4.79 (d, J=15.2 Hz, 1H), 4.56 (s, 1H), 4.32 (dd, J=12.8, 1.2 Hz, 1H), 3.91 (dd, J=12.8, 2.0 Hz, 1H), 3.61 (s, 1H), 3.41 (d, J=2.0 Hz, 1H), 2.06-2.01 (m, 2H), 1.73-1.70 (m, 1H), 1.64-1.61 (m, 1H), 1.20-1.15 (m, 2H).

Example 289 In Vivo Biological Studies

Neuropharmacological Assay (SmartCube™)

In order to demonstrate the utility of the provided compounds to treat neurological and psychiatric diseases and disorders, exemplary compounds were evaluated using the neuropharmacological screen described in S. L. Roberds et al., Front. Neurosci. 2011 Sep. 9; 5:103 (doi: 10.3389/fnins.2011.00103) (“Roberds”). As reported in Roberds, because psychiatric diseases generally result from disorders of cell-cell communication or circuitry, intact systems are useful in detecting improvement in disease-relevant endpoints. These endpoints are typically behavioral in nature, often requiring human observation and interpretation. To facilitate testing of multiple compounds for behavioral effects relevant to psychiatric disease, PsychoGenics, Inc. (Tarrytown, N.Y., “PGI”) developed SmartCube™, an automated system in which behaviors of compound-treated mice are captured by digital video and analyzed with computer algorithms. (D. Brunner et al., Drug Discov. Today 2002, 7:S107-S112). PGI Analytical Systems uses data from SmartCube™ to compare the behavioral signature of a test compound to a database of behavioral signatures obtained using a large set of diverse reference compounds. (The composition of the database as well as validation of the method is further described in Roberds). In this way, the neuropharmacological effects of a test compound can be predicted by similarity to major classes of compounds, such as antipsychotics, anxiolytics and antidepressants.

The SmartCube™ system produces an activity signature indicating the probability that the activity of the test compound at the administered dose matches a given class of neuropharmacological agents. (See, e.g., Roberds, FIGS. 2 and 3). The test compound is simultaneously compared against multiple classes of agents; thus, a separate probability is generated for each behavioral effect measured (e.g., anxiolytic activity, analgesic activity, etc.). In the table below, these probabilities are reported for each behavioral effect measured as follows:

LOQ ≤ + <5%  5% ≤ ++ <25% 25% ≤ +++ <50% 50% ≤ ++++

where LOQ is the limit of quantification.

Provided compounds were dissolved in a mixture of Pharmasolve™ (N-methyl-2-pyrrolidone), polyethylene glycol and propylene glycol, and were injected i.p. 15 min. before the behavioral test. For each compound, injections were administered at 3 different doses. For each behavioral effect measured, results for the most efficacious dose(s) are presented. In the table below, DP: anti-depressant; AX: anxiolytic; SD: sedative hypnotic; PS: anti-psychotic; MS: mood stabilizer; AD: ADHD; CE: cognitive enhancer; AG: analgesic; UN: uncharacterized CNS activity.

The potency of many of the compounds in the table was also determined in the SmartCube™ system. Test compounds were routinely examined at dose levels of 0.3, 1, 3, 10 and 30 mg per kg (mpk), although the dose range was increased or decreased if necessary to obtain a full dose response curve. A compound's minimal effective dose (MED) is a measure of the compounds potency. The MED was defined as the dose (in mpk) having 50% or more total activity in SmartCube. The potencies of the compounds are shown in the table below, with potency values in mpk binned in the following manner:

MED mpk range BIN ≤3 mpk A >3 to 10 mpk B >10 to ≤30 mpk C >30 mpk D Example DP AX SD PS MS AD CE AG UN Potency 1 + ++ ++ ++ + + ++ ++ ++ C 2 + ++ + + + + + ++ +++ C 3 ++ ++ ++ + + + + ++ +++ C 4 ++ +++ ++ ++ + + ++ ++ + C 5 ++ ++ ++ ++ + + ++ ++ +++ A 6 ++ ++ ++ + + + + ++ ++++ B 7 ++ ++ + ++ ++ + ++ + +++ C 8 ++ ++ +++ ++ + + ++ ++ ++ C 9 ++ ++ + ++ + + ++ ++ +++ B 10 ++ ++ ++ +++ + + +++ ++ ++ B 11 ++ + ++ + + + ++ + + C 12 ++ ++ + ++ + + ++ ++ +++ C 13 ++ ++ + ++ + + ++ ++ ++++ B 14 ++ ++ + + + + + ++ ++++ C 15 + + + + + + + + + D 16 + +++ + ++ + + ++ ++ +++ C 17 ++ +++ + ++ + + ++ ++ +++ C 18 ++ ++ ++ ++ + + ++ +++ + C 19 + +++ ++ ++ + + ++ ++ +++ C 20 ++ +++ ++ +++ + + ++ ++ ++ B 21 ++ ++ ++ ++ + + ++ ++ +++ B 22 ++ ++ + ++ + + ++ ++ +++ C 23 ++ +++ + ++ + + ++ ++ ++ C 24 ++ ++ ++ ++ + + + + + C 25 ++ ++ + ++ + + ++ ++ +++ C 26 ++ ++ + ++ + + ++ ++ ++++ A 27 ++ ++ + ++ ++ + ++ ++ +++ B 28 ++ ++ + ++ + + ++ ++ +++ B 29 ++ ++ + + + + + +++ + C 30 + + + + + + + ++ + D 31 ++ ++ + + + + + ++ +++ C 32 ++ ++ ++ ++ + + + ++ + C 33 + ++ + ++ + + ++ ++ +++ C 34 + +++ ++ ++ + + ++ ++ +++ B 35 ++ +++ + ++ + + ++ ++ +++ B 36 ++ ++ ++ ++ + + +++ ++ ++ B 37 + ++ + ++ + + ++ ++ +++ C 38 ++ ++ + ++ + + + ++ +++ C 39 ++ +++ + + + + ++ ++ ++++ B 40 + +++ + ++ + + ++ ++ ++ B 41 ++ ++ + + + + ++ ++ +++ C 42 ++ ++ ++ +++ + + ++ +++ +++ A 43 + +++ ++ ++ + + ++ ++ + C 44 ++ +++ ++ ++ + + ++ ++ +++ B 45 ++ +++ ++ ++ + + ++ ++ ++++ B 46 + ++ ++ + + + ++ ++ ++++ A 47 ++ ++ + ++ ++ + ++ ++ ++++ A 48 + +++ ++ ++ + + +++ ++ ++ A 49 ++ ++ ++ ++ + + ++ ++ ++++ B 50 +++ ++ + ++ + + ++ ++ ++ C 51 ++ ++ + ++ + + ++ ++ ++++ B 52 ++ +++ ++ ++ + + ++ ++ + B 53 ++ ++ ++ ++ + + ++ ++ ++++ B 54 ++ ++ + ++ + + + ++ +++ C 55 + ++ + ++ + + ++ ++ +++ B 56 ++ ++ + + + + ++ +++ + C 57 ++ ++ ++ ++ + + +++ ++ +++ B 58 ++ ++ ++ ++ + + ++ ++ +++ B 59 ++ ++ ++ ++ + + ++ +++ +++ B 60 ++ +++ + ++ + + ++ ++ + C 61 + ++ + ++ + + ++ ++ ++++ C 62 ++ ++ + ++ ++ + +++ ++ ++++ B 63 ++ ++ + + + + ++ ++ ++ C 68 ++ ++ + ++ + + ++ ++ ++ C 72 ++ ++ ++ ++ + + ++ ++ +++ B 73 ++ +++ + ++ + + ++ ++ +++ A 74 ++ ++ + + + + + ++ +++ C 75 + ++ + ++ + + ++ ++ +++ C 76 ++ ++ + ++ + + ++ ++ +++ A 77 ++ ++ + ++ + + ++ ++ +++ A 78 ++ ++ + +++ + + ++ ++ ++++ B 79 ++ +++ ++ ++ + + ++ ++ ++ B 80 + +++ ++ ++ + + + ++ +++ A 81 ++ +++ ++ ++ + + ++ ++ +++ A 82 ++ +++ ++ ++ + + ++ ++ +++ A 83 + ++ + ++ + + ++ ++ ++++ B 84 ++ ++++ + ++ + + ++ ++ +++ B 85 ++ ++ + + + + ++ +++ +++ B 86 + ++ + ++ + + ++ ++ +++ C 87 ++ ++ ++ ++ + + ++ ++ +++ B 88 + +++ ++ ++ + + ++ ++ +++ A 89 + ++ ++ ++ + + ++ ++ ++++ A 90 ++ +++ ++ + + + ++ ++ + C 91 + + ++ ++ + + ++ ++ +++ C 92 ++ ++ + + + + + + ++++ C 93 ++ +++ + ++ + + + ++ + C 94 + ++ + + + + + ++ ++ C 95 ++ ++ + +++ + + ++ ++ +++ A 96 ++ +++ + ++ + + ++ ++ +++ A 97 + ++ ++ ++ + + ++ + ++++ B 98 ++ ++ ++ ++ + + ++ ++ ++++ B 99 ++ +++ + ++ + + ++ ++ +++ B 100 ++ ++ ++ +++ + + ++ ++ +++ A 101 ++ +++ ++ ++ + + + ++ + C 104 ++ ++ ++ ++ + + ++ ++ +++ B 105 ++ ++ + ++ ++ + ++ ++ +++ C 106 + ++ ++ ++ + + ++ + ++++ B 107 + ++ + ++ + + ++ ++ +++ B 111 ++ ++ + ++ + + ++ ++ +++ B 112 ++ ++ + ++ + + +++ ++ + C 113 ++ ++ + ++ + + ++ ++ +++ B 114 ++ ++ + ++ + + ++ ++ +++ C 115 ++ ++ ++ +++ + + ++ ++ +++ B 116 ++ ++ +++ ++ + + ++ ++ +++ B 117 ++ ++ + +++ ++ + ++ ++ + B 118 +++ ++ ++ ++ + + ++ ++ + B 119 ++ +++ ++ ++ + + ++ ++ ++++ A 120 ++ +++ ++ ++ + + ++ ++ ++ B 121 ++ ++ ++ ++ + + ++ ++ +++ A 122 ++ +++ ++ ++ + + +++ ++ +++ A 123 ++ ++ ++ ++ + + + ++ +++ A 124 ++ +++ ++ ++ + + ++ ++ +++ B 125 ++ ++ ++ ++ + + ++ + ++++ A 126 ++ ++ + +++ + + ++ ++ ++++ A 127 ++ ++ + ++ + + ++ ++ +++ A 128 ++ ++ + ++ + + +++ ++ +++ A 129 ++ +++ ++ ++ + + ++ ++ +++ B 130 ++ ++ ++ +++ + + ++ ++ ++++ B 131 ++ ++ ++ ++ ++ + +++ + +++ A 132 ++ +++ ++ ++ + + +++ ++ +++ A 133 ++ ++ + +++ + + ++ ++ ++++ A 134 ++ +++ ++ ++ + + ++ ++ +++ B 135 ++ +++ ++ ++ + + ++ ++ +++ A 136 ++ ++ + ++ + + ++ ++ +++ B 137 + ++ + ++ + + ++ ++ +++ B 138 ++ ++ ++ ++ + + ++ ++ +++ B 139 ++ ++ + ++ + + ++ ++ +++ B 140 ++ ++ + +++ ++ + ++ ++ + C 141 ++ +++ + ++ ++ + ++ ++ +++ B 142 ++ ++ + + + + ++ ++ ++++ B 143 ++ ++ ++ + + + ++ +++ ++ B 144 ++ ++ ++ ++ + + ++ ++ +++ B 145 + ++ + + + + ++ ++ +++ C 146 + +++ + + + + ++ ++ ++ B 147 ++ ++ + + + + + ++ ++++ B 148 ++ ++ ++ ++ ++ + ++ ++ +++ B 149 ++ ++ ++ +++ + + ++ ++ +++ A 150 + ++ + ++ + + + ++ ++++ B 151 ++ +++ ++ ++ + + ++ ++ ++ B 152 ++ +++ + ++ + + ++ ++ +++ A 153 + ++ + ++ + + ++ ++ +++ B 154 + ++ ++ ++ + + ++ ++ ++++ B 155 ++ ++++ ++ ++ + + ++ ++ ++ B 156 + ++ + + + + ++ ++ +++ B 157 ++ ++ + +++ ++ + ++ ++ ++++ A 158 ++ ++ ++ +++ + + ++ ++ ++ B 159 +++ +++ + + + + + + ++ B 160 + ++ + ++ + + ++ ++ +++ A 161 ++ +++ + ++ + + ++ ++ ++ B 162 + +++ + ++ + + ++ ++ +++ B 163 ++ ++ ++ +++ + + ++ ++ +++ B 164 + +++ ++ ++ + + ++ ++ +++ B 165 ++ ++ + ++ + + ++ + ++++ B 166 + ++ ++ ++ + + ++ + ++++ B 167 + ++ + ++ + + + ++ ++++ A 168 ++ ++ + +++ + + ++ + ++++ B 169 ++ ++ ++ ++ + + ++ ++ ++++ A 170 ++ ++ + ++ + + ++ ++ ++++ A 171 ++ +++ ++ ++ + + ++ ++ +++ A 172 + ++ + ++ + + + ++ +++ C 173 + ++ + + + + ++ ++ ++++ B 174 ++ ++ + ++ + + ++ ++ ++++ B 175 ++ ++ + ++ + + ++ ++ ++++ A 176 ++ ++ + ++ + + ++ + ++++ B 177 + ++++ + ++ + + ++ ++ ++++ A 178 ++ ++ ++ ++ + + + ++ +++ C 179 +++ ++ + ++ + + ++ + ++++ A 180 ++ ++ + ++ + + ++ ++ ++++ B 181 + ++ +++ ++ + + ++ ++ ++++ A 182 ++ ++ + +++ + + ++ + ++++ A 183 ++ ++ ++ ++ + + ++ ++ ++++ A 184 ++ ++ + ++ + + ++ + ++++ A 185 + +++ + ++ + + ++ ++ ++++ A 186 ++ ++ + ++ + + ++ ++ +++ B 187 + ++ + +++ + + ++ ++ ++++ A 188 ++ ++ + ++ + + + ++ ++++ A 189 ++ ++ + ++ + + ++ ++ +++ C 190 + ++ ++ ++ + + ++ + ++++ B 191 ++ ++ ++ ++ + + ++ ++ ++++ A 192 + ++ ++ ++ + + ++ + ++++ A 193 + ++ ++ ++ + + ++ ++ ++++ B 194 ++ ++ + ++ + + ++ ++ ++++ B 195 ++ ++ ++ ++ + + ++ ++ ++++ A 196 + ++ + ++ + + ++ + +++ C 197 + ++ + + + + + + ++++ B 198 ++ ++ + +++ + + ++ + +++ A 199 + ++++ ++ ++ + + ++ ++ ++++ A 200 ++ +++ + ++ + + ++ ++ ++++ B 201 + ++ + ++ + + ++ + ++++ B 202 ++ +++ + ++ + + ++ ++ ++++ A 203 ++ ++ ++ +++ + + ++ ++ ++++ B 204 + ++ ++ ++ + + ++ + ++++ B 205 ++ +++ ++ ++ + + ++ + +++ B 206 ++ ++++ + ++ + + ++ ++ +++ B 207 ++ ++ + ++ + + ++ ++ ++++ B 208 ++ +++ + ++ ++ + ++ + +++ B 209 ++ ++ ++ ++ + + ++ +++ +++ B 210 + ++ + + + + ++ ++ ++++ B 211 ++ ++ +++ ++ + + + + ++++ B 212 + ++ + + + + + ++ ++++ C 213 ++ ++ + ++ + + ++ ++ +++ B 214 ++ ++ ++ ++ + ++ ++ ++ ++++ B 215 ++ +++ ++ ++ + + ++ ++ ++++ B 216 ++ ++ + + + + + ++ ++++ B 217 ++ +++ + ++ + + ++ + ++++ C 218 + ++ + ++ + + ++ + ++++ A 219 + ++ + + + + ++ ++ ++++ B 220 + ++++ + + + + ++ ++ ++++ B 221 ++ ++ + ++ + + + + ++++ B 222 + ++ + + + + + + ++++ B 223 ++ ++ ++ + + + + + +++ B 224 + ++ ++ + + + +++ ++ ++ C 225 ++ ++ ++ ++ + + ++ ++ ++++ B 226 ++ ++ + +++ + + ++ + ++ C 227 + ++ + ++ + + + ++ ++++ B 228 ++ ++ + ++ + + + ++ +++ C 229 ++ ++ + ++ + + ++ + +++ B 230 + ++ + ++ + + ++ + ++++ B 231 ++ ++ + + + + ++ ++ +++ C 232 + ++ + + + + + ++ ++ C 233 + ++ + + + + ++ ++ + D 234 + +++ ++ ++ + + ++ ++ ++ C 235 + ++ + + + + + + + D 236 ++ ++ + ++ + + + ++ ++++ B 237 + ++ +++ ++ + + + + ++ C 238 + ++ + ++ + + ++ + ++++ C 239 ++ ++ + ++ + ++ ++ ++ +++ B 240 ++ ++ ++ ++ + + ++ ++ +++ B 248 + ++ + + + + ++ ++ +++ C 249 + ++ + ++ + + ++ ++ +++ B 250 + ++ ++ ++ ++ ++ + + + B 251 + ++ ++ ++ ++ ++ + + ++ B 252 ++ + ++ ++ ++ + + + ++++ A 253 ++ + ++ ++ ++ + + + ++ B 254 + + ++ + + + + + ++ B 255 + + ++ + + + + + ++ C 256 + ++ ++ +++ ++ ++ + + ++ B 257 ++ + +++ ++ ++ + + + ++ B 258 ++ + ++ + ++ + + + + B 259 ++ + +++ + + + + + ++ C 260 + + ++ + ++ ++ + + + B 261 ++++ + ++ + + ++ + + ++ C 262 ++ + ++ + + + + + ++ C 263 + + ++ + + + + + + A 264 ++ ++ ++ +++ ++ ++ + + + B 265 ++ + ++ + + + + + + B 266 + + ++ + ++ + + + + B 267 ++ + ++ + ++ + + + ++ A 268 + ++ ++ +++ ++ ++ + + ++ B 269 ++ + ++ + ++ + + + ++ B 270 ++ ++ ++ + ++++ ++ + + ++ A 271 ++ ++ ++ + +++ ++ + + ++ B 272 + ++ ++ ++ ++ + + + ++ B 273 ++ + +++ + ++ + + ++ ++ A 274 ++ + ++ ++ +++ + + + ++ A 275 + + +++ + ++ + + + ++ B 276 + + ++ + ++ + + + + B 277 + + + + + + + + + D 278 + + + + + + + + + D 279 ++ + ++ ++ +++ + + + ++ B 280 ++ + ++ + ++ + ++ + + B 281 + + + + + + + + + D 282 + + ++ + + + + + + D 283 ++ ++ ++ ++ +++ ++ + + ++ B 284 ++ ++ ++ ++ +++ + + + + C 285 ++ ++ + ++++ ++++ ++ + + ++ B 286 ++ + ++ + ++ + + + ++ C 287 + + ++ + + + + + + D 288 ++ + ++ + + + + + + D

Marble Burying Test

The marble burying assay is used to evaluate the anxiolytic activity of a test compound. After placement in a novel cage containing glass marbles, mice will bury the marbles. Acute administration of anxiolytic agents, as well as antidepressants, decrease this activity at non-sedating doses. Male C57Bl/6J mice from Jackson Laboratories were used in this study and were acclimatized to the experimental room for at least 1 hour prior to testing. Mice (n=10 per group) were administered sterile water or test compound intraperitoneally (i.p.) or orally by gavage (p.o.), or the positive control chlordiazepoxide (15 mg/kg, i.p.), in 10 mL/kg injection volumes, and returned to their home cages for a 30-minute pretreatment period. The mice were then placed individually in clean cages, containing hard wood bedding and twenty black marbles placed in spaced rows of 4×5. After a 30-minute test session, the number of buried marbles was counted. A marble was considered buried if it was pushed at least two thirds into the bedding. Locomotor activity was monitored by measuring the distance traveled using an overhead camera and Video Tracker Software (ViewPoint Life Sciences Software, France). A reduction in the number of marbles buried relative to the vehicle condition indicates an anxiolytic drug-like effect.

Effects of Compounds on Marble Burying in Mice.

Compound Dose Marbles Buried 111  3 mg/kg (p.o.) 10 mg/kg (p.o.) + 30 mg/kg (p.o.) + 123  3 mg/kg (p.o.) 10 mg/kg (p.o.) + 30 mg/kg (p.o.) + 114  1 mg/kg (p.o.)  3 mg/kg (p.o.) + 10 mg/kg (p.o.) + 30 mg/kg (p.o.) + 117  1 mg/kg (p.o.)  3 mg/kg (p.o.) + 10 mg/kg (p.o.) + 30 mg/kg (p.o.) + 118  3 mg/kg (p.o.) 10 mg/kg (p.o.) + 30 mg/kg (p.o.) + 124  3 mg/kg (p.o.) 10 mg/kg (p.o.) + 30 mg/kg (p.o.) + 152  1 mg/kg (i.p.)  3 mg/kg (i.p.) 10 mg/kg (i.p.) + 157  1 mg/kg (i.p.)  3 mg/kg (i.p.) 10 mg/kg (i.p.) + 160  1 mg/kg (i.p.)  3 mg/kg (i.p.) 10 mg/kg (i.p.) + 170  1 mg/kg (i.p.)  3 mg/kg (i.p.) 10 mg/kg (i.p.) + 195  1 mg/kg (i.p.)  3 mg/kg (i.p.) 10 mg/kg (i.p.) + −: No reduction in number of marbles buried +: Significant reduction in number of marbles buried (P < 0.05 vs vehicle)

Stress Induced Hyperthermia Assay

Stress-induced hyperthermia (SIH), mediated by the autonomic nervous system, is well known to occur prior to and during exposure to stress and/or anxiety-inducing situations. In many human anxiety disorders, it occurs as an integral part of the pathology and is often considered a representative symptom of the disease, e.g. in generalized anxiety disorder as defined in DSM-IV. In rodents, acute administration of anxiolytic drugs, such as buspirone or chlordiazepoxide (CDP), are known to reduce the SIH response to stressors such as handling, noise, heat, novelty, or pain. The test involves two measures of rectal temperature repeated in the same animal with a 10-minute interval. On the day prior to testing, male Sprague-Dawley rats from Envigo (n=10 per group) were brought to the experimental room approximately one hour before scheduled lights out and singly housed overnight with food and water ad libitum. On the morning of the experiment, animals were first injected with either vehicle or test compound orally by gavage, or CDP (10 mg/kg, i.p.). One hour following injection, each animal was removed from the holding cage and held in a supine position while the rectal temperature was measured by slowly inserting a rectal probe into the animal's rectum at a length of approximately 0.5 cm. The rectal probe was attached to a PhysiTemp digital thermometer (Fisher Scientific) which provides temperature readings at 0.1° C. accuracy. The probe remained inside the animal for approximately 5 seconds or until body temperature reaches stability. This temperature was recorded as the baseline rectal temperature (T1). The animal was immediately placed back to the holding cage and after a 10-min interval the second rectal temperature (T2) was taken using the same procedure as in measuring T1. Before each insertion, the rectal probe was cleaned with an alcohol pad and lubricated with sterile K-Y jelly. All SIH studies were conducted between 8:00-11:00 a.m. The SIH response was calculated as the difference in temperature between the second and first temperature reading (Delta T, □T). A reduction in □T relative to the vehicle condition indicates an anxiolytic drug-like response.

Effects of Compounds on Stress-induced Hyperthermia (SIH) in Rats.

Dose Compound (mg/kg, PO) Delta T (□T) 152 0.3 mg/kg   1 mg/kg +  3 mg/kg + 10 mg/kg + 30 mg/kg + 157  1 mg/kg  3 mg/kg + 10 mg/kg + 30 mg/kg + 170  3 mg/kg 10 mg/kg + 30 mg/kg + −: No change in Delta T (□T) +: Significant decrease in Delta T (□T) (P < 0.05 vs vehicle)

Amphetamine Induced Hyperlocomotion Assay

Psychostimulants such as amphetamine (AMPH) are frequently used to induce or mimic a manic-like or psychotic-like state. AMPH administration to rodents increases horizontal locomotion, vertical rearing and repetitive stereotyped behaviors, and typical antipsychotic drugs, such as haloperidol, can reduce these behaviors. The antimanic/antipsychotic-like effects of test compounds were evaluated in male C57Bl/6J mice from Jackson Laboratories Mice were acclimatized to the experimental room for at least 1 hour prior to testing. The mice (n=10 per group) were administered vehicle or test compound orally by gavage (10 mL/kg injection volume) and placed individually in the novel open field (OF) chambers (Med Associates) for 30 minutes of baseline activity measurement. Mice were then injected with either water or AMPH (4 mg/kg, i.p., 10 mL/kg) and placed back in the same individual OF chambers for a 60-minute testing session during which the effects of test compounds on stimulant-induced behaviors (total distance traveled, rearing and stereotypy) were measured. A reduction in total distance traveled relative to the vehicle+AMPH condition indicates an antimanic/antipsychotic drug-like response.

Effects of Compounds on AMPH-Induced Hyperlocomotion in Mice.

Total Distance Compound Dose (PO, mg/kg) Traveled (cm) 111  3 mg/kg Compound + AMPH # 10 mg/kg Compound + AMPH 30 mg/kg Compound + AMPH + 123  3 mg/kg Compound + AMPH # 10 mg/kg Compound + AMPH 30 mg/kg Compound + AMPH + 114  3 mg/kg Compound + AMPH 10 mg/kg Compound + AMPH 30 mg/kg Compound + AMPH + 117  3 mg/kg Compound + AMPH 10 mg/kg Compound + AMPH 30 mg/kg Compound + AMPH + 118  3 mg/kg Compound + AMPH 10 mg/kg Compound + AMPH 30 mg/kg Compound + AMPH + 124  3 mg/kg Compound + AMPH 10 mg/kg Compound + AMPH 30 mg/kg Compound + AMPH + #: Significant increase in total distance traveled (P < 0.05 vs vehicle + AMPH group) −: No change in total distance traveled +: Significant decrease in total distance traveled (P < 0.05 vs vehicle + AMPH group)

Tail Suspension Test

The tail suspension test (TST) is a rodent screening test for potential (human) antidepressant drugs. It is based on the assumption that an animal will actively try to escape an aversive (stressful) stimulus. If escape is impossible, the animal will eventually stop trying (“give up”). In the TST, a mouse is suspended by the tail so that its body dangles in the air, head downward. Mice initially struggle to face upward and climb to a solid surface. When the animal stops struggling and hangs immobile it is considered to have “given up”. Shorter periods of immobility are characteristic of antidepressant-like activity. Accordingly, longer periods of immobility are considered indicative of a depressive-like state. It has been shown that treatment with an antidepressant drug will decrease the time the animal spends immobile. See generally L. Steru et al., Psychopharmacology (Berl). 1985; 85(3):367-70; B. Thierry et al., Psychopharmacology 1986; 90:284-85.

Procedure.

Adult male AJ mice from Jackson Laboratories received vehicle (sterile water) or test compound orally by gavage, or the positive control desipramine (20 mg/kg, i.p.), in 10 mL/kg injection volumes, 30 min before being subjected to the Tail Suspension Test. In this test, mice are placed in the Tail Suspension chambers (white polyvinylchloride cubicles measuring 33×33×31.75 cm Med Associates, Inc. St. Albans, Vt.) by a piece of transparent (Scotch®) tape attached to the tail, from about the mid-tail, with approximately 2 cm of tape past the end of the tail for 10 min during which the time spent immobile is measured. A reduction in total time immobile relative to the vehicle condition indicates an antidepressant drug-like response.

Effects of Compounds in a Tail-Suspension Test (TST) in Mice.

Dose (PO, Mean Total Time Compound mg/kg) Immobile (sec) 111  3 mg/kg 10 mg/kg 30 mg/kg 123  3 mg/kg 10 mg/kg 30 mg/kg 114  3 mg/kg 10 mg/kg 30 mg/kg + 117  3 mg/kg 10 mg/kg + 30 mg/kg 118  3 mg/kg 10 mg/kg 30 mg/kg 124  3 mg/kg 10 mg/kg 30 mg/kg −: No decrease in Mean Total Time Immobile +: Significant decrease in Mean Total Time Immobile (P < 0.05 vs vehicle)

Prepulse Inhibition of Startle

The acoustic startle is an unconditioned reflex response to an external auditory stimulus. Prepulse inhibition of startle (PPI) refers to the reduction in the startle response caused by the presentation of a low-intensity auditory stimulus prior to the startle stimulus. The PPI paradigm is used for the study of schizophrenia and antipsychotic action due to the similarities between the results from human and rodent studies. PPI has been used as a tool for the assessment of deficiencies in sensory-motor gating observed in schizophrenia and to screen for potential antipsychotic drugs. Various psychotomimetic drugs, such as phencyclidine (PCP), can disrupt PPI. In mice, antipsychotic drugs such as haloperidol can increase PPI and clozapine can reverse the disruption of PPI induced by PCP.

Male C57Bl/6J mice from Jackson Laboratories were placed in the PPI chambers (Med Associates) for a 5 min session of white noise (70 dB) habituation. After the habituation period, the test session was initiated. The session started with a habituation block of 6 presentations of the startle stimulus alone, followed by 10 PPI blocks, each of which consisted of 6 different types of trials. Trial types were: ‘null’ (no stimuli), ‘startle’ (120 dB), ‘startle plus prepulse’ (4, 8, and 12 dB over background noise, i.e., 74, 78, or 82 dB) and ‘prepulse’ (82 dB). Trial types were presented in a random order within each block. Each trial started with a 50 ms stimulus-free period during which baseline movements were recorded. This was followed by a subsequent 20 ms period during which the prepulse stimulus was presented and responses to the prepulse measured. After a further 100 ms period, the startle stimulus was presented for 40 ms and responses recorded for 100 ms from startle onset. Responses were sampled every ms. The inter-trial interval was variable with an average of 15 s (range from 10 to 20 s). In ‘startle’ trials, the basic auditory startle response was measured. The basic startle response was calculated as the mean startle response of all ‘startle’ trials (i.e., excluding the first habituation block). In ‘startle plus prepulse’ trials, the degree of inhibition of the normal startle was calculated and expressed as a percentage of the basic startle response.

Mice were treated with vehicle or test compound orally by gavage, or haloperidol (1 mg/kg, i.p.), in 10 mL/kg injection volumes, 30 min prior to PPI test. The PPI enclosures were cleaned following each test. An increase in the percent PPI relative to the vehicle condition indicates an antipsychotic drug-like response.

Effects of Compounds on Pre-Pulse Inhibition (PPI) in Mice.

Dose (PO, Compound mg/kg) Effect 111  3 mg/kg 10 mg/kg 30 mg/kg +++ 123  3 mg/kg 10 mg/kg 30 mg/kg 114  3 mg/kg 10 mg/kg 30 mg/kg 117  3 mg/kg 10 mg/kg 30 mg/kg +++ 118  3 mg/kg 10 mg/kg 30 mg/kg 124  3 mg/kg 10 mg/kg 30 mg/kg −: No change in PPI +: Significant increase in PPI at one pre-pulse intensity (P < 0.05 vs vehicle) ++: Significant increase in PPI at two pre-pulse intensities (P < 0.05 vs vehicle) +++: Significant increase in PPI at three pre-pulse intensities (P < 0.05 vs vehicle)

In Vivo Microdialysis of the Prefrontal Cortex and Nucleus Accumbens

In vivo microdialysis is a biological sampling technique used to evaluate the extracellular concentrations of neurotransmitters in specific brain regions following administration of a test compound to animals. Dysfunctions of brain neurotransmitter systems, such as dopamine (DA), norepinephrine (NE), serotonin (5-HT) and/or acetylcholine (ACh), have been linked to cognitive deficits (e.g., memory impairment, inattention, etc.), which may be observed in human aging, Alzheimer's disease (AD) and Attention Deficit Hyperactivity Disorder (ADHD). Drugs that increase extracellular concentrations of DA, NE, 5-HT and/or ACh in the prefrontal cortex (PFC), such as donepezil and methylphenidate, can reverse cognitive deficits in humans and animals.

Adult male C57Bl/6 mice (n=6 per group) from Jackson Laboratories were anesthetized using isoflurane (2%, 800 mL/min O2). Bupivacaine/epinephrine was used for local anesthesia and carprofen was used for peri-/post-operative analgesia. The animals were placed in a stereotaxic frame (Kopf instruments, USA) and I-shaped microdialysis probes (polyacrylonitril membrane, BrainLink, the Netherlands) were inserted into the PFC (2 mm exposed membrane; coordinates for the tips of the probes: posterior (AP)=+2.0 mm to bregma, lateral (L)=−0.5 mm to midline and ventral (V)=−3.3 mm to dura, the toothbar set at 0.0 mm) and the NAcc (coordinates for the tips of the probes: posterior (AP)=+0.8 mm to bregma, lateral (L)=+0.9 mm to midline and ventral (V)=−4.2 mm to dura, the toothbar set at 0.0 mm (Paxinos and Franklin, 2001)). After surgery, animals were housed individually in cages and provided food and water ad libitum. In vivo microdialysis experiments were performed one day after surgery. On the day of the experiment, the microdialysis probes were connected with flexible PEEK tubing to a microperfusion pump (Harvard PHD 2000 Syringe pump, Holliston, Mass. or similar). Microdialysis probes were perfused with artificial cerebrospinal fluid (aCSF) containing 147 mM NaCl, 3.0 mM KCl, 1.2 mM CaCl2 and 1.2 mM MgCl2, at a flow rate of 1.5 L/min. Microdialysis samples were collected for 30 minute periods by an automated fraction collector (820 Microsampler, Univentor, Malta or similar) into polystyrene mini-vials already containing 15 μL of 0.02 M formic acid (FA) and 0.04% ascorbic acid in ultrapurified H2O. Four basal samples were collected before the administration of vehicle or test compound (i.p., 5 mL/kg injection volumes). Samples were collected for an additional 240 min following vehicle or compound administration. All the dialysis samples were stored at −80° C. awaiting analysis. After the experiment, the mice were sacrificed and brain tissue was collected for probe verification. Each dialysis sample was analyzed for concentrations of DA, NE, 5-HT and ACh as determined by HPLC with tandem mass spectrometry (MS/MS) detection using labeled isotopes of the analytes of interest as internal standards. Data was expressed as a percentage of basal output, calculated by dividing each post-dose time point with the average basal output=100%. An increase in the post-dose percent basal output of DA, NE, 5-HT and/or ACh in the PFC relative to the vehicle condition indicates a potential cognition-enhancing drug-like response. Graphical representations of the observed changes in neurotransmitter concentrations are shown in the indicated Figures.

Effects of Compounds on Neurotransmitters in the PFC and NAcc in Mice.

Dose PFC PFC PFC PFC (mg/kg, [DA] [5-HT] [NE] [ACh] Compound IP) (FIG. 1) (FIG. 2) (FIG. 3) (FIG. 4) 152 10 mg/kg + + + + 157 30 mg/kg + + + 170 30 mg/kg + + Dose NAcc NAcc NAcc NAcc (mg/kg, [DA] [5-HT] [NE] [ACh] Compound IP) (FIG. 5) (FIG. 6) (FIG. 7) (FIG. 8) 152 10 mg/kg + + 157 30 mg/kg + 170 30 mg/kg + −: No change in extracellular concentration (P < 0.05 vs vehicle) +: Significant increase in extracellular concentration (P < 0.05 vs vehicle)

LIST OF ABBREVIATIONS

5-HT 5-hydroxytrytophan (serotonin)

ACh acetylcholine

ACN acetonitrile

aCSF artificial cerebrospinal fluid

AMPH Amphetamine

AP anterior-posterior

° C. degrees Celsius

CDP chlordiazepoxide

cm centimeter

DA dopamine

FA formic acid

g gram

HPLC high performance liquid chromatography

IP intraperitoneal

L lateral

LC liquid chromatography

μL microliter

MD microdialysis

mg/kg milligram per kilogram

mL milliliter

mm millimeter

mM millimolar

min/Min minute

MRM multiple reaction monitoring

MS mass spectrometry

MSq mean squared

NAcc nucleus accumbens

NE norepinephrine

nM nanomolar

P p-value

PCP Phencyclidine

PPI Pre-pulse Inhibition

% percentage

PFC prefrontal cortex

pmol picomole

PSIG pounds per square inch gage

QC quality control

sec seconds

SEM standard error of the mean

SIH stress-induced hyperthermia

TST tail suspension test

V ventral

UP ultra-pure

It may be found upon examination that additional species and genera not presently excluded from the claims to pharmaceutical compositions and chemical compounds are not patentable to the inventors in this application. In that case, the subsequent exclusion of species and genera in applicants' claims are to be considered artifacts of patent prosecution and not reflective of the inventors' concept or description of their invention. The invention, in a composition aspect, is all compounds of formula I except those that are in the public's possession.

It is to be understood that the invention is not limited to the particular embodiments of the invention described above, as variations of the particular embodiments may be made and still fall within the scope of the appended claims.

Claims

1. A composition comprising a compound, wherein said compound is:

or a pharmaceutically acceptable salt thereof, and wherein said composition is greater than 90% enantiomerically pure.

2. The composition according to claim 1, wherein said composition is greater than 95% enantiomerically pure.

3. A composition comprising a compound, wherein said compound is:

or a pharmaceutically acceptable salt thereof, and wherein said composition is greater than 90% enantiomerically pure.

4. The composition according to claim 3, wherein said composition is greater than 95% enantiomerically pure.

5. A pharmaceutical composition comprising a compound, wherein said compound is:

or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier, adjuvant, or vehicle.

6. A pharmaceutical composition comprising a compound, wherein said compound is:

or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier, adjuvant, or vehicle.

7. The pharmaceutical composition of claim 6, wherein said compound is:

or a pharmaceutically acceptable salt thereof.

8. The pharmaceutical composition of claim 5, wherein said compound is:

or a pharmaceutically acceptable salt thereof.

9. The pharmaceutical composition of claim 5, wherein said compound is:

or a pharmaceutically acceptable salt thereof.

10. The pharmaceutical composition of claim 5, wherein said compound is:

or a pharmaceutically acceptable salt thereof.

11. The pharmaceutical composition of claim 5, wherein said compound is:

or a pharmaceutically acceptable salt thereof.

12. A method for alleviating or inhibiting the progress of a neurological or psychiatric disease or disorder selected from the group consisting of depression, bipolar disorder, pain, schizophrenia, obsessive compulsive disorder, addiction, social disorder, attention deficit hyperactivity disorder, an anxiety disorder, a movement disorder, epilepsy, Alzheimer's disease, Parkinson's disease, autism and cognitive impairments in a subject in need thereof, comprising administering to said subject an effective amount of a compound, wherein said compound is:

or a pharmaceutically acceptable salt thereof.

13. The method of claim 12, wherein said neurological or psychiatric disorder is depression.

14. The method of claim 13, wherein the depression is treatment-resistant depression (TRD), major depressive disorder (MDD), unipolar depression, bipolar depression or depression associated with another disease or disorder.

15. The method of claim 12, wherein said neurological or psychiatric disorder is bipolar disorder.

16. The method of claim 12, wherein said neurological or psychiatric disorder is pain.

17. The method of claim 12, wherein said neurological or psychiatric disorder is schizophrenia.

18. The method of claim 12, wherein said neurological or psychiatric disorder is obsessive compulsive disorder.

19. The method of claim 12, wherein said neurological or psychiatric disorder is addiction.

20. The method of claim 12, wherein said neurological or psychiatric disorder is social disorder.

21. The method of claim 12, wherein said neurological or psychiatric disorder is attention deficit hyperactivity disorder.

22. The method of claim 12, wherein said neurological or psychiatric disorder is an anxiety disorder.

23. The method of claim 12, wherein said neurological or psychiatric disorder is a movement disorder.

24. The method of claim 12, wherein said neurological or psychiatric disorder is Alzheimer's disease.

25. The method of claim 12, wherein said neurological or psychiatric disorder is Parkinson's disease.

26. The method of claim 12, wherein said neurological or psychiatric disorder is autism.

27. The method of claim 12, wherein said neurological or psychiatric disorder is cognitive impairments.

28. A method of alleviating or inhibiting the progress of a neurological or psychiatric disease or disorder selected from the group consisting of depression, bipolar disorder, pain, schizophrenia, obsessive compulsive disorder, addiction, social disorder, attention deficit hyperactivity disorder, an anxiety disorder, a movement disorder, epilepsy, Alzheimer's disease, Parkinson's disease, autism and cognitive impairments in a subject in need thereof, comprising administering to said subject an effective amount of a compound, wherein said compound is:

or a pharmaceutically acceptable salt thereof.

29. The method of claim 28, wherein said neurological or psychiatric disorder is depression.

30. The method of claim 29, wherein the depression is treatment-resistant depression (TRD), major depressive disorder (MDD), unipolar depression, bipolar depression or depression associated with another disease or disorder.

31. The method of claim 28, wherein said neurological or psychiatric disorder is bipolar disorder.

32. The method of claim 28, wherein said neurological or psychiatric disorder is pain.

33. The method of claim 28, wherein said neurological or psychiatric disorder is schizophrenia.

34. The method of claim 28, wherein said neurological or psychiatric disorder is obsessive compulsive disorder.

35. The method of claim 28, wherein said neurological or psychiatric disorder is addiction.

36. The method of claim 28, wherein said neurological or psychiatric disorder is social disorder.

37. The method of claim 28, wherein said neurological or psychiatric disorder is attention deficit hyperactivity disorder.

38. The method of claim 28, wherein said neurological or psychiatric disorder is an anxiety disorder.

39. The method of claim 28, wherein said neurological or psychiatric disorder is a movement disorder.

40. The method of claim 28, wherein said neurological or psychiatric disorder is Alzheimer's disease.

41. The method of claim 28, wherein said neurological or psychiatric disorder is Parkinson's disease.

42. The method of claim 28, wherein said neurological or psychiatric disorder is autism.

43. The method of claim 28, wherein said neurological or psychiatric disorder is cognitive impairments.

44. The method of claim 12, wherein said neurological or psychiatric disorder is epilepsy.

45. The method of claim 28, wherein said neurological or psychiatric disorder is epilepsy.

Referenced Cited
U.S. Patent Documents
3438995 April 1969 Faust et al.
3470179 September 1969 Ott
3536809 October 1970 Applezweig
3549624 December 1970 Conover et al.
3551427 December 1970 Ott
3598123 August 1971 Zaffaroni
3845770 November 1974 Theeuwes et al.
3916899 November 1975 Theeuwes et al.
3995052 November 30, 1976 Jirkovsky et al.
4008719 February 22, 1977 Theeuwes et al.
4021451 May 3, 1977 Dobson et al.
4021452 May 3, 1977 Floyd
4036842 July 19, 1977 Dobson et al.
4041169 August 9, 1977 Demerson et al.
4066648 January 3, 1978 Oka et al.
4127665 November 28, 1978 Sarges et al.
4337343 June 29, 1982 Maillard et al.
4500543 February 19, 1985 Debernardis et al.
4556656 December 3, 1985 McAll
4904300 February 27, 1990 Schneider
4963568 October 16, 1990 Schoenleber et al.
4994486 February 19, 1991 Schoenleber et al.
4999359 March 12, 1991 Vecchietti et al.
5032598 July 16, 1991 Baldwin et al.
5041451 August 20, 1991 Colle et al.
5059595 October 22, 1991 Le Grazie
5073543 December 17, 1991 Marshall et al.
5109008 April 28, 1992 Scopes et al.
5120548 June 9, 1992 McClelland et al.
5134127 July 28, 1992 Stella et al.
5288749 February 22, 1994 Meyer et al.
5304657 April 19, 1994 Toki et al.
5354556 October 11, 1994 Sparks et al.
5391556 February 21, 1995 Heckel et al.
5393759 February 28, 1995 Combourieu et al.
5464834 November 7, 1995 Peligion et al.
5532203 July 2, 1996 Fory et al.
5532233 July 2, 1996 Weber et al.
5591767 January 7, 1997 Mohr et al.
5621133 April 15, 1997 Deninno et al.
5639476 June 17, 1997 Oshlack et al.
5646173 July 8, 1997 Bos et al.
5656658 August 12, 1997 Hammarberg et al.
5674533 October 7, 1997 Santus et al.
5684020 November 4, 1997 Peligion et al.
5733566 March 31, 1998 Lewis
6031099 February 29, 2000 Moltzen et al.
6235774 May 22, 2001 Fagrig et al.
6262044 July 17, 2001 Møller et al.
6313309 November 6, 2001 Baxter et al.
6436943 August 20, 2002 Stoltefuss et al.
6503913 January 7, 2003 Goldmann et al.
7019026 March 28, 2006 Andersen et al.
7282499 October 16, 2007 Arjona et al.
7297704 November 20, 2007 Sabb et al.
7414068 August 19, 2008 Lim et al.
7544717 June 9, 2009 Hom et al.
7745462 June 29, 2010 Fairhurst et al.
7884109 February 8, 2011 Ohlmeyer et al.
8227625 July 24, 2012 Corbera-Arjona et al.
8710245 April 29, 2014 Shao et al.
9216975 December 22, 2015 Napoletano et al.
9351954 May 31, 2016 Shao et al.
10085968 October 2, 2018 Shao et al.
10196403 February 5, 2019 Hanania et al.
10336732 July 2, 2019 Xie et al.
20030073687 April 17, 2003 Bebbington et al.
20030149057 August 7, 2003 Want et al.
20040180883 September 16, 2004 Gilmore et al.
20040220402 November 4, 2004 Chow et al.
20050032873 February 10, 2005 Hatzenbuhler et al.
20050075366 April 7, 2005 Heidelbaugh et al.
20050187281 August 25, 2005 Hinze et al.
20050239832 October 27, 2005 John et al.
20050267199 December 1, 2005 Hom et al.
20060047127 March 2, 2006 Arjona
20060148872 July 6, 2006 Chow et al.
20060258714 November 16, 2006 Heffernan et al.
20070032481 February 8, 2007 Dvorak et al.
20070072926 March 29, 2007 Chow et al.
20070154534 July 5, 2007 Sheitman et al.
20070185144 August 9, 2007 Zhong et al.
20080081910 April 3, 2008 Saab et al.
20080113961 May 15, 2008 Nishi et al.
20080255239 October 16, 2008 Chow et al.
20080306082 December 11, 2008 Dahnke et al.
20090069305 March 12, 2009 Gaul et al.
20090118283 May 7, 2009 Defert et al.
20090318690 December 24, 2009 Sasaki et al.
20100035887 February 11, 2010 Ricciardi
20100178299 July 15, 2010 Sitkovsky et al.
20100197714 August 5, 2010 Wunsch et al.
20100295881 November 25, 2010 Yao et al.
20120171199 July 5, 2012 Dobson et al.
20130109677 May 2, 2013 Shao et al.
20140256712 September 11, 2014 Shao et al.
20150031709 January 29, 2015 Campbell et al.
20160083399 March 24, 2016 Shao et al.
20160264597 September 15, 2016 Chytil et al.
20170001987 January 5, 2017 Xie et al.
20180030064 February 1, 2018 Xie et al.
20180057506 March 1, 2018 Chytil et al.
20180093974 April 5, 2018 Xie et al.
20180118727 May 3, 2018 Campbell et al.
20190038594 February 7, 2019 Bowen et al.
20190256525 August 22, 2019 Bauer et al.
20190308990 October 10, 2019 Xie et al.
20200129477 April 30, 2020 Hopkins et al.
20200179336 June 11, 2020 Hopkins
Foreign Patent Documents
2010325925 June 2011 AU
2016200448 February 2016 AU
2031684 June 1991 CA
1072411 May 1993 CN
1300291 June 2001 CN
1860112 November 2006 CN
100384824 April 2008 CN
101468986 July 2009 CN
101468987 July 2009 CN
101759710 June 2010 CN
102731574 October 2012 CN
3827727 February 1990 DE
368175 May 1990 EP
416740 March 1991 EP
0431421 June 1991 EP
984365 February 1965 GB
01006267 January 1989 JP
H2243691 September 1990 JP
H 03163068 July 1991 JP
H 03223277 October 1991 JP
049367 January 1992 JP
03223277 October 2001 JP
2002512233 April 2002 JP
2002512244 April 2002 JP
2003261566 September 2003 JP
2004269449 September 2004 JP
2005145859 June 2005 JP
2006117568 May 2006 JP
2014214130 November 2014 JP
2015510513 April 2015 JP
WO 91/08205 June 1991 WO
WO 9203434 March 1992 WO
WO 1992003434 March 1992 WO
WO 92/14465 September 1992 WO
WO 92/15592 September 1992 WO
WO 94/00441 January 1994 WO
WO 96/04287 February 1996 WO
WO 96/38435 December 1996 WO
WO 99/01437 January 1999 WO
WO 99/46237 September 1999 WO
WO 99/46267 September 1999 WO
WO 0000487 January 2000 WO
WO 2000/023445 April 2000 WO
WO 2000/035915 June 2000 WO
WO 2000/043397 July 2000 WO
WO 2000/068230 November 2000 WO
2000078742 December 2000 WO
WO 2000/078742 December 2000 WO
WO 2001/017516 March 2001 WO
WO 2001/19831 March 2001 WO
WO 2001/0132610 May 2001 WO
WO 2001/0132655 May 2001 WO
WO 2001032610 May 2001 WO
WO 2001032655 May 2001 WO
WO 2001062233 August 2001 WO
WO 2001072745 October 2001 WO
WO 2002/012189 February 2002 WO
WO 2002/022614 March 2002 WO
WO 2002/02066443 August 2002 WO
WO 2001080893 October 2002 WO
WO 2002083667 October 2002 WO
WO 2002/02102387 December 2002 WO
WO 2003/006455 January 2003 WO
WO 2003/03035065 May 2003 WO
WO 2003092374 November 2003 WO
WO 2004/004726 January 2004 WO
WO 2004024081 March 2004 WO
WO 2004/035812 April 2004 WO
WO 2004/066912 August 2004 WO
WO 2004/078723 September 2004 WO
WO 2004/082687 September 2004 WO
WO 2004/087680 October 2004 WO
WO 2004089913 October 2004 WO
WO 2004/112719 December 2004 WO
WO 2005012291 February 2005 WO
WO 2005/035518 April 2005 WO
WO 2005/072412 August 2005 WO
WO 2005/073236 August 2005 WO
WO 2005/079800 September 2005 WO
WO 2005/087779 September 2005 WO
WO 2005087714 September 2005 WO
WO 2005095326 October 2005 WO
WO 2005111025 November 2005 WO
WO 2006/014135 February 2006 WO
WO 2006/014136 February 2006 WO
WO 2006/015259 February 2006 WO
WO 2006/030124 March 2006 WO
WO 2006/053274 May 2006 WO
WO 2006066172 June 2006 WO
WO 2006089053 August 2006 WO
WO 2006/117305 November 2006 WO
WO 2007/001939 January 2007 WO
WO 2007/002681 January 2007 WO
WO 2007/006546 January 2007 WO
WO 2007/095586 August 2007 WO
WO 2007/102999 September 2007 WO
WO 2007/120594 October 2007 WO
WO 2007/126041 November 2007 WO
WO 2008011560 January 2008 WO
WO 2008/042422 April 2008 WO
WO 2008/048981 April 2008 WO
WO 2008/058342 May 2008 WO
WO 2008/119689 October 2008 WO
WO 2008/125348 October 2008 WO
WO 2008/155132 December 2008 WO
WO 2009/009550 January 2009 WO
WO 2009/057974 May 2009 WO
WO 2009/067202 May 2009 WO
WO 2009/068467 June 2009 WO
WO 2009/072621 June 2009 WO
WO 2009/085256 July 2009 WO
WO 2010/053583 May 2010 WO
WO 2010/092180 August 2010 WO
WO 2010/092181 August 2010 WO
WO 2010090716 August 2010 WO
WO 2010093425 August 2010 WO
WO 2011017389 February 2011 WO
WO 2011/036889 March 2011 WO
WO 2011/060035 May 2011 WO
WO 2011/060217 May 2011 WO
WO 2011/069063 June 2011 WO
WO 2011/081205 July 2011 WO
WO 2011/133729 October 2011 WO
WO 2012/020133 February 2012 WO
WO 2012/122340 September 2012 WO
WO 2013/010453 January 2013 WO
WO 2013050334 April 2013 WO
WO 2013/067248 May 2013 WO
WO 2013091773 June 2013 WO
WO 2013119895 August 2013 WO
WO 2013/192346 December 2013 WO
2014078454 May 2014 WO
WO 2014/078454 May 2014 WO
WO 2014/106238 July 2014 WO
WO 2006/066950 June 2016 WO
WO 2018/023072 February 2018 WO
WO 2011/094740 August 2018 WO
WO 2018151861 August 2018 WO
WO 2019161236 August 2019 WO
WO 2019161238 August 2019 WO
WO 2020118032 June 2020 WO
9102744 February 1992 ZA
Other references
  • Akdemir, A., et al. “Identification of novel α7 nicotinic receptor ligands by in silico screening against the crystal structure of a chimeric α7 receptor ligand binding domain.” Bioorganic and Medicinal Chemistry. (2012), vol. 20, pp. 5992-6002 (Year: 2012).
  • Anxiety and Depression Association of America. “Depression.” Accessed Mar. 16, 2019. (Jan. 29, 2009). Available from: < https://adaa.org/understanding-anxiety/depression/treatment >. (Year: 2009).
  • National Institute of Mental Health. “Bipolar Disorder.” (Apr. 2016). Accessed Mar. 16, 2019. Available from: < https://www.nimh.nih.gov/health/topics/bipolar-disorder/index.shtml >. (Year: 2016).
  • Nemade, R., et al. “Schizophrenia Medication Treatment Options.” (Feb. 15, 2006). Accessed Mar. 16, 2019. Available from: < https://www.mentalhelp.net/articles/schizophrenia-medication-treatment-options/ >. (Year: 2006).
  • National Institute of Mental Health. “Obsessive-Compulsive Disorder.” (Jan. 2016). Accessed Mar. 16, 2019. Available from: < https://www.nimh.nih.gov/health/topics/obsessive-compulsive-disorder-ocd/index.shtml >. (Year: 2016).
  • CDC. “Treatment of ADHD.” (Jan. 30, 2016). Accessed Mar. 16, 2019. Available from: < https://www.cdc.gov/ncbddd/adhd/treatment.html >. (Year: 2016).
  • “Find the Best Epilepsy Treatment for You.” (May 23, 2015). Accessed Mar. 16, 2019. Available from: < https://share.upmc.com/2015/05/epilepsy-treatment/ >. (Year: 2016).
  • Wilkinson, D., et al. “Cholinesterase Inhibitors Used in the Treatment of Alzheimer's Disease.” Drugs Aging (2004), vol. 21, Issue 7, pp. 453-478. (Year: 2004).
  • Werber, E.A., et al. “The beneficial effect of cholinesterase inhibitors on patients suffering from Parkinson's disease and dementia.” J. Neural Transm. (2001), vol. 108, pp. 1319-1325. (Year: 2001).
  • Berge, S., et al. “Pharmaceutical Salts.” Journal of Pharmaceutical Sciences. (Jan. 1977), vol. 66, No. 1, pp. 1-19 (Year: 1977).
  • Nguyen, Lien, Ai., et al. “Chiral Drugs: An Overview.” Int. J. Biomed Sci. (Jun. 2006), vol. 2, Issue 2, pp. 85-100. (Year: 2006).
  • Bianchi, et al, “Model studies towards stephaoxocanes: Construction of the 2-oxa-4-azaphenalene core of stephaoxocanidine and eletefine”, European Journal of Organic Chemistry (2003), (24), 4731-4736.
  • Ram et al., “Synthesis and structure-activity relationships of 1-substituted-aminomethyl-3-phenyl/methyl-1,3-dihydroisobenzofurans and 4-substituted-amino-1-phenyl/methylisochromans—a new class of antihistaminics”, Indian Journal of Chemistry, Section B: Organic Chemistry Including Medicinal Chemistry (1984), 23B(12), 1261-7.
  • Rekka et al., “Structural features of some diphenhydramine analogs that determine the interaction with rat liver cytochrome P-450”, Agents and Actions (1989), 27(1-2), 184-7.
  • The list of search results of CAPLUS, Apr. 21, 2016. (Total 20 pages.)
  • Answer Summary, from the search of CAPLUS, Apr. 21, 2016 (Total 29 pages.)
  • International Search Report and Written Opinion, dated Dec. 21, 2017, in the corresponding PCT Appl. No. PCT/US17/44511.
  • Ahmad, I and Snieckus, V., “A Convenient Entro into the Rhoeadan Skelton. Total Synthesis of (±)-cis-alpinigenine”, Canadian Journal of Chemistry, 60(12):2678-2686 (1982).
  • American Chemical Society, STN Database RN 63463-05-8 entered Nov. 16, 1984.
  • Antoz, F.J., et al., “The Structure and Chemistry of Actinobolin”, Journal of American Chemical Society, 92(16):4933-4942 (1970).
  • AU Application No. 2013216935, Examination Report No. 2 dated Aug. 1, 2017.
  • Bakshi, et al., “Antagonism of Phencyclidine-Induced Deficits in Prepulse Inhibition by the Putative Atypical Antipsychotic Olanzapine,” Psychopharamcology, 122(2):198-201, 1995.
  • Berardi, et al., “4-(Tetralin-1-yl)-and-4-(Naphthalen-1yl)akyl] Derivatives of 1-Cyclohexylpiperazine as σ Receptor Ligands with Agonists σ2 Activity”, Journal of Medicinal Chemistry, American Chemical Society, 47(9):2308-23-17 (2004).
  • Berardi, et al., “A Multireceptorial Binding Reinvestigation on an Extended Class of σ Ligands: N-[ω-(Indan-1-yl and Tetralin-1-yl)alkyl] Derivatives of 3,3-Dimethylpiperdine Reveal High Affinities Towards σ1 and EBP Sites”, Bioorganic & Medicinal Chemistry, 9(5):1325-1335 (2001).
  • Berardi, et al., “Novel Potent σ1 Ligands: N-[ω-(Tetralin-1-yl)alkyl] piperdine Derivatives”, Journal of Medicinal Chemistry, American Chemical Society, 38(21):4255-4260 (1996).
  • Berge, S.M., et al., “Pharmaceutical Salts”, Journal of Pharmaceutical Sciences, 66(1):1-19 (1977).
  • Boger, D.L., et al., “Thermal Cycloaddition of 1,3,5-Triazine with Enamines: Regiospecific Pyrimidine Annulation”, Journal of Organic Chemistry, 47:2673-2675 (1982).
  • Böhme, H. And Hitzel, V., “Homoisochroman-Derivative mit basischer Seitenkette in 1-Stellung”, Archiv der Pharmazie 306:948-953 (1973) No English Translation.
  • Böhme, H. and Ziegler, F., “The Aminomethylation of 1-cyano-isochromane and 1-cyano-isothiochromane”, Arch Pharm (Weinheim), 307(4):287-290 (1974) with English Translation.
  • CAPULUS Search Results of Apr. 6, 2016; 102 pages.
  • CAS Database Regisry 444792-99-8 (XP-002742897) Aug. 24, 2002; 1 page.
  • CAS Database Regisry 444793-00-4 (XP-002742898) Aug. 24 ,2002; 1 page.
  • CAS Database Regisry 444796-01-5 (XP-002742896) Aug. 24, 2002; 1 page.
  • CAS Database Regisry 46490-93-1 (XP-002742899) Nov. 16, 1984; 1 page.
  • CAS Database Regisry 738532-48-4 (XP-002742900) Sep. 3, 2004; 1 page.
  • CAS Database Regisry Accession No. 1022058-43-0, May 23, 2008.
  • CAS Database Regisry Accession No. 1022339-80-5, May 25, 2008.
  • CAS Database Regisry Accession No. 1022468-83-2, May 25, 2008.
  • CAS Database Regisry Accession No. 1022813-67-7, May 27, 2008.
  • CAS Database Regisry Accession No. 1023480-64-9, May 29, 2008.
  • CAS Database Regisry Accession No. 1024262-27-8 Jun. 1, 2008.
  • CAS Database Regisry Accession No. 3549452-84-9, Sep. 28, 2001.
  • CAS Database Regisry Accession No. 3594552-83-8, Sep. 28, 2001.
  • CAS Database Registry Accession Nos. 131022-75-8, 1310059-007-4, 1310059-06-3, 1310059-08-5 and 1310059-09-6 as cited in the Japanese Office Action dated Mar. 14, 2017 for Japanese Application No. 2014-556702.
  • CAS Database Registry Accession No. 340968-07-2, Jun. 14, 2001.
  • CAS Database Registry Accession No. 359452-60-1, Sep. 28, 2001.
  • CAS Database Registry No. 1027177-28-1 Jun. 11, 2008.
  • CAS Database Registry No. 1935196-69-2 Jun. 20, 2016.
  • CAS Registry No. 724648-33-5; STN entry date Sep. 10, 2004; Chemical name: Quinazoline, 4-benzo[b]thien-2-yl-1,4-dihydro- (cited Aug. 1, 2017, obtained from https://scifinder.cas.org on Aug. 10, 2017).
  • CAS Registry No. 736880-30-1; STN entry date Sep. 1, 2004; Chemical name: Quinazoline, 4-benzo[b]thien-2-yl-1,4-dihydro-2-methyl- (cited Aug. 1, 2017, obtained from https://scifinder.cas.org on Aug. 10, 2017).
  • CAS Registry No. 775528-08-0; STN entry date Nov. 7, 2004; Chemical name: Quinazoline, 4-benzo[b]thien-2-yl-1,4-dihydro-2-methyl (cited Aug. 1, 2017, obtained from https://scifinder.cas.org on Aug. 10, 2017).
  • CAS Registry No. 790156-85-3; STN entry date Nov. 28, 2004; Chemical name: Quinazoline, 4-benzo[b]thien-2-yl-1,4-dihydro- (cited Aug. 1, 2017, obtained from https://scifinder.cas.org on Aug. 10, 2017).
  • CAS Registry No. 933704-221-3, Apr. 30, 2007.
  • CAS Registry No. 1541037-08-04 date unknown.
  • Chemical Abstracts STN Registry Database, record for RN 1784628-34-7, Entered into STN on Jun. 19, 2015. (Year: 2015).
  • Chihara, et al., “Preparation of Benzothiiophene Derivatives as Blood Platelt Aggregation Inhibitors”, Retrieved from STN Database Asccession No. 1992:128652 and JP03223277a, Yoshitomi Pharmacetuical Industries Ltd., Oct. 2, 1991.
  • CN Office Action in Application No. 201410333332.1, dated Nov. 2, 2015 with translation.
  • Corbera, et al., “A Medicinal-Chemistry-Guided Approach to Selective and Druglike Sigma 1 Ligands”, ChemMedChem, 1(1):140-154 (2006).
  • Dammacco, M., et al., “Lithiation of N-Alkyl-(o-totyl)aziridine: Stereoselective Synthesis of Isochromans”, Journal of Organic Chemistry, 74:6319-6322 with supplemental material pp. S1-S34 (2009).
  • Debernadis, J.F., et. al., “Conformationally Defined Adrenergic Agents. 4. 1-(Aminomethyl)phthalans: Synthesis and Pharmacological Consequences of the Phtalan Ring Oxygen Atom”, Journal of Medicinal Chemistry, 30:178-184 (1987).
  • Dehaven-Hudkins, et al., “Characterization of the Binding of [3H](+)pentazocine to σ Recognition Sites in Guinea Pig Brain”, European Journal of Pharmacology—Molecular Pharmacology Section 227:371-378 (1992).
  • Deninno, M.P., et al., “Synthesis and Dopaminergic Activity of 3-Substituted 1- (Aminomethyl)-3,4-dihydro-5,6-dihydroxy-1H-2-benzopyrans: Characterization of an Auxiliary Binding Region in the D1 Receptor”, Journal of Medicinal Chemistry, 34:2561-2569 (1991).
  • Disabled World Towards Tomorrow, “Neurological Disorders: Types, Research & Treatment” URL: https://www.disabled-world.com/health/neurology downloaded on Nov. 1, 2017; 5 pages.
  • Dobson, et al., “Pyrano Heterocycles I. The Synthesese of Isochromans and the Novel Thieno[3,2-c]pyran, Benzothieno[3,2-c]pyran, and Pyrano[4,3-b]benxofuran Systems”, Journal of Hetercyclic Chemistry, 12(3):591-594, 1975, 4 pages.
  • Ellis, “Affective Disorders (Mood Disorders)”, Healthline Part 1 of 7 Overview; URL: http://www.healthline.com/health/affective-disorders, 5 pages, downloaded Jul. 25, 2015.
  • Emedicine Health, “Brain Cancer: Get Facts on Treatment, Causes and Symptoms”, URL: https://www,emedicinehealth.com/brain_cancer/article_em.htm?pf=2; 15 pages downloaded 2015.
  • EP Application No. 123747266.8, Communication Pursuant to Article 94(63) EPC dated Dec. 21, 2017.
  • EP Application No. 13747266.8, Partial Supplementary European Search Report dated Aug. 14, 2015; 11 pages.
  • EP Application No. 10835185.9, Extended European Search Report dated Apr. 4, 2013, 15 pages.
  • EP Patent Application No. 13747266.8, Communication Pursuant to Article 94(3) dated Nov. 18, 2016.
  • Gaur,S., et al. “CoMFA and CoMSIA Studies on a set of Benzyl Piperazines, Piperadines, Pyrazinopyridoindoles, Pyrazinoisoquinolines and Semi Rigid analogs of Diphenydramine”, Medicinal Chemistry Research, 13(8-9):746-757 (2004).
  • Ghaemi, et al., “Does Olanzapine have Antidepressant Properties? A Retrospective Preliminary Study”, Bipolar Disorders, 2:196-199, 2000.
  • Ghasemi, et al., “The Role of NMDA Receptors in the Pathophysiology and Treatment of Mood Disorders”, Neuroscience and Biobehavioral Reviews, 47:336-358, 2014.
  • Girke, W.P.K., “Elektrophile Aromatische Substitutionsreaktionen Mit Protonierten 1,3- Diazinen II. Darstellung and Eigenschaften 4-arylsubstituierter 3,4-Dichrochinazolin-Derivate”, European Journal of Inorganic Chemistry, 112(4):1348-1358 (1979) [English Abstract and machine translation of entire referenc- 24 pages].
  • Gleason, et al., “Blockade of Phencyclidine-Induced Hyperlocomotion by Olanzapine, Clozapine and Serotonin Receptor Subtype Selective Antagnoists in Mice”, Psychopharmacology, 129:79-84, 1997.
  • Gould, P.L., “Sale Selection for Basic Drugs”, International Journal of Pharmaceutics, 33:201-217 (1986).
  • Grilliot, A-L and Hart, D.J., “Guanidinium Carboxylates: Preparation of 3-Carboxyoctahydro-9aH-Pyrimidin-9a-Ylium Chloride”, Hetercoycles, 39(2):435-438 (1994).
  • Gronowitz, et al, The Reaction of 5-Bromo- and 2-Bromopyrimidine with Organolithium Compounds, Acta Chemica Scandinavica 19(7):8 pages (1965).
  • Hanner, et al., “Purification Molecular Cloning, and Expression of the Mammalian Sigma1-Binding Site”, Proc. Natl. Aca. Sci., 93:8072-8077 (1996).
  • Hayakawa, K., et al., “Addition Reactions of (Phenylsulfonyl)propadiene with 1-Pryrrolidinyl Enamines of Cyclic Ketones: Syntheses and Reactions of 1,3-Dienes Possessing an Allyl Sulfone Moiety”, Journal of Organic Chemistry, 51:5100-5105 (1986).
  • Hejl, et al., “Prepulse Inhibition in Patients with Alzheimer's Disease”, Neurobiology of Aging, 25:1045-1050 (2004).
  • Hörig, H. and Pullman, W., “From Bendch to Clinic and Back: Perspective on the 1st IQPC Translational Research Conference”, Journal of Translational Medicine, 2:44, 8 pages (2004).
  • Huang, N-A, et al., “Thiation Reactions of Some Active Carbonyl Compoounds with Sulfur Transfer Reagents”, The Journal of Organic Chemistry, 52(2):169-172 (1987).
  • Ingebrigsten, T., et al., “Palladium-Catalysed Synthesis of Pyrimidines”, Heterocycles, 65(11):2593-2603 (2005).
  • International Application No. PCT/US2010/05884, International Search Report and Written Opinion dated Aug. 25, 2011, 10 pages.
  • International Application No. PCT/US2013/025260, International Preliminary Report on Patentability issued by the International Searching Authority, dated Aug. 21, 2014 (10 pages).
  • International Application No. PCT/US2013/025260, International Search Report and Written Opinion issued by the International Searching Authority, dated Apr. 17, 2013 (10 pages).
  • International Application No. PCT/US2016/017527 International Search Report dated Apr. 4, 2016.
  • International Application No. PCT/US2016/017539, International Search Report dated Apr. 21, 2016.
  • International Application No. PCT/US2016/017539, Written Opinion dated Apr. 21, 2016.
  • International Application No. PCT/US2017/044517, International Search Report and Written Opinion dated Jan. 11, 2018.
  • International Application No. PCT/US2018/044854, International Search Report and Written Opinion dated Apr. 10, 2018, 13 pages.
  • Ito, N., et al., “A Medium-Term Rat Live Bioassy for Rapid in vivo Dection of Carcinogenic Potential of Chemicals”, Cancer Science, 94(1):3-8 (2003).
  • Jacobs, et al., “1-Imidizolyl(alkyl)-Substituted Di- and Tetrahydroquinolines and Analogues: Syntheses and Evaluation fo Dual Inhibitors of Thromboxane A2 Synthase and Aromatase”, Journal of Medicinal Chemistry, 43(9):1841-1851, 2000.
  • Jaskowska, J. and Kowalski, P., “N-Alkylation of Imides Using Phase Transfer Catalysts Under Solvent-Free Conditions”, Journal Heterocyclic Chemistry, 45:1371-1375 (2008).
  • Jentsch, et al., “The Neuropsychopharmacology of Phencyclidine: From NMDA Receptor Hypfunction to the Dopamine Hypothesis of Schizophrenia”, Neurpsychopharmacology, 20(3):201-225, 1999.
  • JP Application No. 2012-542219, Office Action dated Nov. 21, 2014, 9 pages including translation.
  • JP Application No. 2014-556702 , Notice of Reasons for Rejection dated Jul. 19, 2016 (with translation).
  • JP Application No. 2014-556702 , Notice of Reasons for Rejection dated Mar. 14, 2017 (with translation).
  • Kapur, et al., “NMDA Receptor Antagonists Ketamine and PCP Have Direct Effects on the Dopamine D2 and Serotonin 5-HT2 Receptors—Implications for Models of Schizophrenia”, Molecular Psychiatry, 7:837-844, 2002.
  • Karran, et al., “The Amyloid Cascade Hypothesis for Alzheimer's Disease: An Appraisal for the Development of Therapeutics”, Nature, 10:698-712 (2011).
  • Katsuki, et al., “Excitotoxic Degeneration of Hypthalamic Orexin Neurons in Slice Culture”, Neurobiology of Disease, 15:61-69, 2004.
  • Kornev, et al. CAS STN Abstrack , publ 2009 RN 1202851-83-9.
  • Kostin, et al., “Lack of Hypocretin Attenuates Behavioral Changes Produced by Glutamatergic Activation of the Perifornical-Lateral Hypthalamic Area”, Sleep, 37(5):1011-1020, 2014.
  • Krogsgaard-Larsen, et al., Texbook of Drug Design and Discovery Madsen, U. (Ed.). (2009). Textbook of Drug Design and Discovery, Fourth Edition. Boca Raton: CRC Press. (2002).
  • Kumar, A., et al., “Catecholamines in a Semi-Rigid Framework: Synthesis & Biological Activity of N-Substituted I-Aminomethyl-5,6- & 6,7-dihydroxyisochromans”, Indian Journal of Chemistry 26B:47-51 (1987).
  • Kumar, A., et al., “Phenethylamine in a Semi-Rigid Framework: Synthesis & Biological Activity of N-Substituted I-Aminomethyl-5,6- & 6,7-dimethoxyisochromans”, Indian Journal of Chemistry 16B:793-796 (1978).
  • Langa,et al., “Generation and Phenotypic Analysis of Sigma Receptor type 1 (σ1) Knockout Mice”, European Journal of Neuroscience, 18:2188-2196 (2003).
  • Lima, et al., Bioisosterism: A Useful Strategy for Molecular Modification and Drug Design, Current Medicinal Chemistry, 12:23-49 (2005).
  • Lindvall, O. and Kokaia, Z., “Stem Cells for the Treatment of Neurological Disorders”, Nature, 441:1094-1096 (2006).
  • Lowry, et al., “Protein Measurement with the Folin Phenol Reagent”, Journal Biochemistry, 193:265 (1951).
  • Macchia, B., et al., “Conformationally Restrained Analogs of Sympathomimetic Catecholamines, Synthesis Conformational Analysis, and Adrenergic Activity of Isochroman Derivatives”, Journal of Medicinal Chemistry, 36:3077-3086 (1993).
  • Maier, et al., “Novel Spiropiperdines as Highly Potent and Subtype Selective σ-Receptor Ligands. Part 1”, Journal of Medicinal Chemistry, 45:438-448 (2002), Journal Medicinal Chemistry, 45:4923-4930 (2002).
  • Maier, et al., “Novel a Receptor Ligands, Part 2. SAR of Spiro[[2]benxopyran-1,4-piperdines] and Spiro [[2]benzofuran-1,4′-piperidines] with Carbon Substituents in Position 3”, Journal Medicinal Chemistry, 45:4923-4930 (2002).
  • Marcus, et al., “The Efficacy and Safety of Aripiprazole as Adjunctive Therapy in Major Depressive Disorder. A Second Multicenter, Randomized, Double-Blind, Placebo-Controlled Study”, Journal of Clinical Psybhopharmacology, 28(2):156-165, 2008.
  • Mashkovskiy, Drugs, Moscow, New Wave, LLC, vol. 1, p. 11 (2002) with translation.
  • Mayo Clinic Symptoms and Causes, “Seasonal Affective Disorder (SAD)”, URL: https://www.mayoclinic.org/diseases-conditions/seasonal-affective-disorder/syymptoms-causes; 2 pages, downloaded 2015.
  • Mokrosz, et al., “Structure-Activity Relationship Studies of CNS Agents. Part 14:3 Structural Requirements for the 5-HT1A and 5-HT2A Receptor of Simple 1-(2-pyrimidinyl)piperazine Derivatives”, Pharmazie, 49(H11) 6 pages (1994).
  • Moreno, et al., “Preclinical Models of Antipsychotic Drug Action”, International Journal of Neuropsychopharmacology, 16:2131-2144, 2013.
  • Movassaghi, M. and Hill, M.D., “Single-Step Synthesis of Pyrimidine Derivatives”, Journal of American Chemical Society, 128:14254-14255 (2006).
  • MX Application No. MX/a/2012/006326, Examination Report dated Jul. 4, 2013, with English translation, 6 pages.
  • Nakashima, T., et al., “Regulation of Folding and Photochromic Reactivity of Teraylenes Through a Host-Guest Interaction”, Chem. European Journal, 17:10951-10957 (2011).
  • Nishimura, Y., et al., “Syntheses and Activities of some Bactobolin Derivatives”, Journal of Antibiotics, 45(5):735-741 (1992).
  • Nordquist, et al., “Effects of Aripiprazole/OPC-14597 on Motor Activity, Pharmacological Models of Psychosis, and Brain Activity in Rats”, Neuropharmacology, 54:405-416, 2008.
  • NZ Application No. 600008, Firstt Examination Report dated Mar. 11, 2013 in NZ , 3 pages.
  • NZ Application No. 626068 Examination Report dated Oct. 8, 2015, 3 pages.
  • NZ Application No. 711802 Examination Report dated Oct. 8, 2015, 5 pages.
  • Papillion, J.P.N., et al., “Structure-Activity Relationships, Pharmacokinetics, and in Vivo Activity of CYP11B2 and CYP11B1 Inhibitors”, Journal of Medicinal Chemistry, 58:4749-4770 (2015).
  • Pittenger, et al., “The NMDA Receptor as a Therapeutic Target in Major Depressive Disorder”, CNS & Neuroological Distorders—Drug Targets, 6(2): 101-115, 2007.
  • PUBCHEM CID 1215079, create date Feb. 7, 2007, p. 3 compound; accessed Nov. 13, 2017; 9 pages.
  • Quirion, et al., “A Proposal for the Classification of Sigma Binding Sites”, Trends in Pharamcology Science, 13:85-86 (1992).
  • Quiroz, T., et al., “A Practical Method for the Synthesis of Pyrrolizidine, Indolizidine and Pyrroloazepinolizidine Nucleus”, Tetrahedron Letters, 48:1571-1575 (2007).
  • Radesca, et al. , “Synthesis and Receptor Binding of Enantiomeric N-Substituted cis-N-[2-(3,4-Dichlorophenyl)ethyl]-1-(1-pyrrolidinyl) cyclohexylamines as High-Affinity σ Receptor Ligands”, Journal of Medicinal Chemistry, 34:3058-3065 (1991).
  • Ross, L.O., et al., “Potential Anticancer Agents. XVIII. Synthesis of Substituted 4,5-Trimethylenepyrimidines”, Journal of American Chemical Society, 84(12):3108-3114 (1959).
  • Sakai, et al., “Facile and Efficient Synthesis of Polyfunctionalized Benzofurans: Three-Component Coupling Reactions from an Alkynylsilane, and o-Hydroxybenzaldehyde Derivative, and a Secondary Amine by a Cu(1)-Cu(II) Cooperative Calatytic System”, RwreHWSEON IWRRWEA, 49:3437-3440 (2008).
  • Salomone, A., et al., “Preparation of Polysubstituted Isochromanes by Addition of ortho-Lithiated Aryloxiranes to Enaminones”, Journal of Organic Chemistry, 78:11059-11065 (2013).
  • Saxena, M., et al., “Synthesis of some Substituted Pyrazinopyridoindoles and 3D QSAR Stuies along with Related Compounds: Piperazines, Piperidines, Pyrazinoisoquinolines. And Diphenhydramine, and its Semi-Rigid Analogs as Antihistamines (H1)”, Bioorganic & Medicinal Chemistry 14:8249-8258 (2006).
  • Schäfer, S., “Failure is an Option: Learning from Unsuccessful Proof-of-Concept Trials”, Drug Discovery Today, 13(21/22):913-916 (2008).
  • Schmitz, et al., “Hippocampal Neuron Loss Exceeds Amyloid Plaque Load in a Transgenic Mouse Model of Alzheimer's Disease”, American Journal of Pathology, 164(4):1045-1050 (2004).
  • Schow, et al., “Novel Sigma Receptor Ligands 2.”, Bioorganic & Medicinal Chemistry Letters, 3(2):221-224 (1993).
  • SG Application No. 201204089-5, Written Opinion dated Sep. 20, 2013, 12 pages.
  • Singapore Application No. 10201401661, Search Report and Written Opinion dated Jun. 15, 2015, 10 pages.
  • Snyder, et al., “Receptor Mechanisms in Antipsychotic Drug Action: Focus on Sigma Receptors”, Journal of Neuropsychiatry, 1(1):7-15 (1989).
  • Steliou, K., et al., “Group 14 Metal Assisted Carbon-Sulfur Bond Formation”, Journal of Organic Chemistry, 50(24):4969-4971 (1985).
  • Strekowski, et al., “Synthesis of 2-Chloro-4,6-di(heteraryl)pyrimidines”, Journal of Heterocyclic Chemistry, 27:1393-1400 (1989).
  • Swerdlow et al., “Seroquel Restores Sensorimotor Gating in Phencyclidine-Treated Rats,” Journal of Pharmacology and Experimental Therapeutics, vol. 279, No. 3, pp. 1290-1299, Dec. 1996.
  • Toffano, M., et al., “Asymmetric Routes Towards Polyfunctionalized Pyrrolidines: Application to the Synthesis of Alkaloid Analogues”, Tetrahedron: Asymmetry, 14:3365-3370 (2003).
  • Torrado, et al., “Novel Selective and Potent 5-HT Reuptake Inhibitors with 5-HT1D Antagonist Activity: Chemistry and Pharmacological Evaluation of a Series of Thienopyran Derivatives”, Bioorganic & Medicinal Chemistry, 12(20):5277-5295, 2004, 19 pages.
  • Trehan, “A New Synthesis of 13-aza-18-nor-17oxo-A-nor-3-thiaestra-1,5(10), 9(11)- triene” Retrieved from STN Database Accession No. 1986:225089 & Indian Journal of Chemistry, Section 6: Organic Chemistry Including Medicinal Chemistry, 24B(6):659-661 (1985).
  • Trehan, “Synthesis of 2,3,13-Triaza-18-nor-17-oxo-3-phenyl-A-nor-estra-1,5(10), 9(11)triene & 2,3,13-Triaza-7,7-dimethy1-18-nor-17-oxo-3-phenyl-A-nor-estra-1,5(10), 9(11)-triene”, Indian Journal of Chemistry, 19B:243-245 (1980).
  • Van Der Stoel, et al., “Di-TT-methane Regarrangement of 4-Heteroaryl-1,4(or 3,4)-dihydropyrimidines”, Journal of the Chemical Society, Perkin Transactions 1, 4 pages Nov. 2, 1978.
  • Vecchietti, et al.,“(1S)-1-(Aminomethyl)-2-(arylacetyl)-1,2,3,4-tetrahydroisoquinoline and Heterocycle-Condensed Tetrahydropyridine Derivatives: Members of a Novel Class of Very Potent K Opiod Analgesics”, Journal of Medicinal Chemistry, 34(8):2624-2633 1991.
  • Walker, et al., “Sigma Receptors: Biology and Function”, Phamacological Reviews, 42(4):355-402 (1990).
  • Weis, et al., “The Crystal and Molecular Structure of 4,6,6-trimethyl-2-phenyl-1,6- dihydropyrimidine”, Hetercycles, 19(3):6 pages (1982).
  • Williams, M., et al., “Emerging Molecular Approaches to Pain Therapy”, Journal of Medicinal Chemistry, 42(9):1481-1500 (1999).
  • Winhusen, T.M., et al., “A Placebo-Controlled Screening Trial of Tiagabine, Sertraline and Donepezil as Cocaine Dependence Treatments”, Addiction, 100(Suppl.1):68-77 (2005).
  • Xi, Z., et al., Preparation of Partially Substituted 1-Halo- and 1,4-Dihalo-1,3-Dienes via Reagent-Controlled Desilyation of Halogenated 1,3-Dienes, Journal of Organic Chemistry, 71:3154-3158 (2006).
  • US Food and Drug Administration, “Highlights of Prescribing Information: Abilify,” FDA label, last revised Dec. 2014, 84 pages.
  • International Application No. PCT/US2017/044511, International Preliminary Report on Patentability issued by the International Searching Authority, dated Jan. 29, 2019, 8 pages.
  • International Application No. PCT/US2017/044517, International Preliminary Report on Patentability issued by the International Searching Authority, dated Jan. 29, 2019, 6 pages.
  • CAS Registry No. 1027834-86-1, Entered STN: Jun. 13, 2008, 4 pages.
  • CAS Registry No. 1071058-54-2, Entered STN: Nov. 6, 2008, 4 pages.
  • CAS Registry No. 40196-92-7, Entered STN: Nov. 16, 1984, 2 pages.
  • CAS Registry No. 40196-93-8, Entered STN: Nov. 16, 1984, 2 pages.
  • Datta et al., “Studies in Sulphur Heterocycles. Part 5. Further Use of 6,7-Dihydribenzo[b]thiphen-4[5H]-one in the Synthesis of Substituted Benzo[b]thiophene Derivatives”, J. Chem. Research (S), 1988, 72-73.
  • Davis et al., “Benzothiophene Containing Rho Kinase Inhibitors: Efficacy in an Animal Model of Glaucoma”, Bioorganic & Medicinal Chemistry Letters, Jun. 1, 2010, 20(11):3361-3366.
  • Devani et al., “Synthesis of 2-Aminothiophenes & Thieno[2,3-d]pyrimidines”, Indian Journal of Chemistry, May 1976, 14B:357-360.
  • Extended European Search Report in EP Appln. No. 17835384.3, dated Mar. 9, 2020, 10 pages.
  • Frohlich et al., “A Novel Synthesis of 3,3-(Spiro)Substituted Azetidines”, Heterocycles, 1994, 37(3):1897-1891.
  • Google.com [online] “Parkinson's Disease—Symptoms, Diagnosis and Treatment.” Jan. 22, 2006, [Retrieved on Dec. 28, 2018] Retrieved from URL <https://www.google.com/search?q=Parkinson+disease+treatment&source=Int&tbs=cdr%3A1%2Ccd_max%3A2%2F8%2F2012&tbm=>, 2 pages.
  • Hopkinsmedicine.org [online] “Treatment for Tourette Syndrome: Johns Hopkins Pediatric Neurology,” Apr. 2006, [Retrieved on Dec. 28, 2018] Retrieved from URL <https://www.hopkinsmedicine.org/neurology_neurosurgery/centers_clinics/pediatric-neurology/conditions/tourettes_syndrome/treatment.html>, 1 page.
  • International Preliminary Report on Patentability in PCT Appln. No. PCT/US2018/044854, dated Feb. 4, 2020, 8 pages.
  • Mayoclinic.org [online] “Fibromyalgia Treatment: Is Neurontin Effective?” Jul. 2009, [Retrieved Oct. 18, 2019], Retrieved from URL <https://www.mayoclinic.org/diseases-conditions/fibromyalgia/expert-answers/fibromyalgia-treatment/faq-20058273>, 3 pages.
  • Medicinenet.com [online] “Alzheimer's Disease Treatment, Symptoms, Stages & Life Expectancy.” Jul. 2007, [Retrieved on Dec. 28, 2018], Retrieved from URL <https://www.medicinenet.com/alzheimers_disease_causes_stages_and_symptoms/article.htm#alzheimers_disease_medications>, 16 pages.
  • Medlineplus.gov [online] “Symptoms, Diagnosis and Treatment: Alzheimer's Disease.” Fall 2010, [Retrieved on Dec. 28, 2018], Retrieved from URL <https://medlineplus.gov/magazine/issues/fall10/articles/fall10pg19.html>, 5(3):19.
  • Michaeljfox.org [online] “Parkinson's Disease,” May 2007, [Retrieved Dec. 28, 2018] Retrieved from URL <https://www.michaeljfox.org/understanding-parkinsons/living-with-pd/topic.php?causes>, 5 pages.
  • Ng, et al., “Design and synthesis of hydroxyethylamine (HEA) BACE-1 inhibitors: prime side chromane-containing inhibitors,” Bioorganic & Medicinal Chemistry Letters, 2013, 23(16):4674-4679.
  • PUBCHEM CID 4878038, create date Sep. 17, 2005, accessed Feb. 22, 2019; 12 pages.
  • PUBCHEM CID 4878041, create date Sep. 17, 2005, accessed Feb. 22, 2019; 14 pages.
  • Ross et al., “α2 Adrenoceptor Agonists as Potential Analgesic Agents. 2. Discovery of 4-(4-Imidazo)-1,3-dimethyl-6,7-dihydro-thianaphthene as a High-Affmity Ligand for the α2D Adrenergic Receptor”, J. Med. Chem., 2000, 43:1423-1426.
  • Shklyaeva et al., “2-Amino-6-(3,4-ethylenedioxythiophen-2-yl)-4-(2-thienyl)-pyrimidine: Synthesis and Properties”, Russian Journal of Organic Chemistry, 2010, 46(6):938-940.
  • Sridhar et al., “Synthesis and Anticancer Activity of Some Novel Pyrimidine Derivatives”, International Journal of Pharmaceutical Sciences and Research, Sep. 29, 2011, 2(10):2562-2565.
  • Stanetty et al., “Heterocyclische Spiroverbindungen Spiroverbindungen: Spiro [benzo[b]thiophen-4(5H),3′-pyrrolidine]”, Arch. Pharm., 1984, 317:168-176 With English Abstract.
  • Togna, et al., “1-Phenil-6, 7-dihydroxy-isochroman inhibits inflammatory activation of microglia,” Brain Research Bulletin, 2013, 95:33-39.
  • Caira “Crystalline Polymorphism of Organic Compounds,” Topics in Current Chemistry, 1998, 163-208.
  • Correll et al. “Safety and Effectiveness of SEP-363856 in Schizophrenia: Results of a 6-Month, Open-Label Extension Study”, American College of Neuropsychopharmacology, Poster. Dec. 2019, 1 page.
  • Dedic et al. “SEP-363856, A Novel Psychotropic Agent With a Unique, Non-D2 Receptor Mechanism of Action”, The Journal of Pharmacology and Experimental Therapeutics, vol. 371, Pages 1-14. Oct. 2019.
  • Dedic et al. “The Novel, Non-D2 Psychotropic Agent SEP-363856 Modulates Presynaptic Dopamine Function in Mice”, American College of Neuropsychopharmacology, Poster. Dec. 9, 2019, 1 page.
  • Fujima et al. “Synthesis of (s)-3-(N-Methylamino)-1-(2-thienyl)propan-1-ol: Revisiting Eli Lilly's Resolution-Racemization-Recycle Synthesis of Duloxetine for it's Robust Processes”, Organic Process Research and Development, 2006, 10(5):905-913.
  • Hopkins et al. “Effects of SEP-363856 on Negative Symptoms in Schizophrenia: Analysis of an Acute, Placebo-Controlled Trial of a Novel Psychotropic Agent With No Dopamine-D2/5-Ht2a Antagonist Activity”, American College of Neuropsychopharmacology, Poster. Dec. 2019, 1 page.
  • INTL Application No. PCT/US2018/000078 International Search Report and Written Opinion, dated May 25, 2018, 9 pages.
  • INTL Application No. PCT/US2019/018263 International Search Report and Written Opinion, dated Apr. 2, 2019, 8 pages.
  • INTL Application No. PCT/US2019/018265 International Search Report and Written Opinion dated May 29, 2019, 17 pages.
  • INTL Application No. PCT/US2019/064646, International Search Report and Written Opinion, dated Mar. 9, 2020, 13 pages.
  • Jones et al. “SEP-0363856, A Novel Psychotropic Agent With a Unique, Non-D2 Mechanism of Action”, European College of Neuropsychopharmacology Congress, Poster. Sep. 2019.
  • Kay et al., “The positive and negative syndrome scale (PANSS) for schizophrenia,” Schizophrenia Bulletin, 1987, 13(2):261-276.
  • Koblan et al. “A Non-D2-Receptor-Binding Drug for the Treatment of Schizophrenia”, The New England Journal of Medicine, Apr. 16, 2020, 382(16):1497-1506.
  • Koblan et al. “A Phase 1 Open Label Safety and Tolerability Study of SEP-363856, A Novel Non-D2 Mechanism of Action Molecule, in Patients With Schizophrenia”, American College of Neuropsychopharmacology, Poster. Dec. 2016, 1 page.
  • Koblan et al. “Efficacy and Safety of SEP-363856 in the Treatment of Schizophrenia: A 4-Week, Randomized, Placebo-Controlled Trial of a Novel Compound With a Non-D2 Mechanism of Action”, American College of Neuropsychopharmacology Annual Meeting, Poster. Dec. 2018, 1 page.
  • Koblan et al. “Efficacy and Safety of SEP-363856, A Novel Non-D2 Agent, in the Treatment of Schizophrenia: A 4-Week, Randomized, Placebo-Controlled Trial”, European College of Neuropsychopharmacology Congress, Poster. Sep. 2019, 1 page.
  • Koblan et al. “Efficacy and Safety of SEP-363856, A Novel Psychotropic Agent With a Non-D2 Mechanism of Action, in the Treatment of Schizophrenia: A 4-Week, Randomized, Placebo-Controlled Trial”, American Psychiatric Association, Poster. May 2019, 1 page.
  • Koblan et al. “Efficacy and Safety of SEP-363856, A Novel Psychotropic Agent With a Non-D2 Mechanism of Action, in the Treatment of Schizophrenia: A 4-Week, Randomized, Placebo-Controlled Trial”, American Society of Clinical Psychopharmacology, Poster. May 2019, 1 page.
  • Koblan et al. “Efficacy and Safety of SEP-363856, A Novel Psychotropic Agent With a Non-D2 Mechanism of Action, in the Treatment of Schizophrenia: A 4-Week, Randomized, Placebo-Controlled Trial”, US Psychiatric and Mental Health Congress, Poster. Oct. 2019, 1 page.
  • Koblan et al. “Efficacy and Safety of SEP-363856, A Novel Psychotropic Agent With a Non-D2 Mechanism of Action, in the Treatment of Schizophrenia: A Randomized, Placebo-Controlled Trial”, Neuroscience Education Institute, Poster. Nov. 2019, 1 page.
  • Koblan et al. “SEP-363856, A Novel Psychotropic Agent With a Non-D2 Mechanism of Action for the Treatment of Schizophrenia”, The 2019 Congress of the Schizophrenia International Research Society, Oral Presentation. 20 pages. Apr. 2019.
  • Koblan, “SEP-363856, A Candidate Antipsychotic and Antidepressant Compound With a Novel Non-D2 Mechanism of Action”, 6th Biennial Schizophrenia International Research Society Conference, Oral Presentation. 31 pages. Apr. 2018.
  • Mahableshwarkar et al., “Replication of a statistical method to reduce pseudospecificity and enhance understanding of score changes among PANSS factors,” Presented at the International Society of CNS Clinical Trials and Methodology, Paris, France, Aug. 31-Sep. 2, 2017, 1 page.
  • Poola et al. “Pharmacokinetics, Safety, and Tolerability of SEP-363856 in Healthy Adult Male Subjects and in Adult Patients With Schizophrenia Following Oral Administration”, American College of Clinical Pharmacology, Poster. Sep. 2018, 1 page.
Patent History
Patent number: 11077090
Type: Grant
Filed: Jul 28, 2017
Date of Patent: Aug 3, 2021
Patent Publication Number: 20180028492
Assignees: Sunovion Pharmaceuticals Inc. (Marlborough), PGI Drug Discovery LLC (Paramus, NJ)
Inventors: Vadim Alexandrov (Tarrytown, NY), Carrie A. Bowen (Uxbridge, MA), Taleen G. Hanania (Valhalla, NY), Noel Aaron Powell (Westford, MA)
Primary Examiner: John S Kenyon
Application Number: 15/663,672
Classifications
Current U.S. Class: Additional Hetero Ring Containing (504/225)
International Classification: C07D 311/76 (20060101); A61K 31/353 (20060101); A61K 31/13 (20060101); C12P 17/06 (20060101); C07D 313/08 (20060101); A61K 45/06 (20060101); A61K 31/352 (20060101);